1
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
2
|
Deng Y, Zhong L, Zhao Y, Wan P, Zhang Y, Liao Y, Zhang H, Wang M, Liu B. A Dendrobium chrysotoxum extract erianin induce AML cells death by activating PPARɑ and downregulating PI3K/AKT signaling pathways. Toxicon 2025; 261:108371. [PMID: 40280444 DOI: 10.1016/j.toxicon.2025.108371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Erianin is a biphenyl compound with low toxicity and a single structure that is extracted from Dendrobium officinale. The wide spectrum of pharmacological properties and excellent toxicity of erianin have been comprehensively proven in multiple tumors. However, less is known about the toxicity of erianin in acute myeloid leukemia (acute myeloid leukemia AML). Here, we explored the anti-AML capacity and potential mechanisms of erianin. Cells proliferation and cytotoxicity of AML cells of erianin was detected by CCK-8 assay and flow cytometer was conducted to assess AML cells apoptosis rate. Erianin blocked the AML cells cycle at the G2/M phase by regulating cell cycle-related protein and P21, P27, and P53 mRNA expression. Additionally, we first filtered PPARɑ and PIK3R1 through network pharmacology, protein-protein interaction (PPI) network, and GO and KEGG pathway enrichment analysis and confirmed their binding with erianin by molecular docking analysis.The cellular thermal shift assay (CETSA) and the drug affinity responsive target stability assay (DARTS) further verified that PPARɑ was an effective target of erianin. Specifically, erianin was found to inhibit the transcriptional level of PIK3R1 by promoting the protein expression of PPARɑ, thereby inhibiting the PI3K/AKT pathway. The inhibitory effect of erianin was partially neutralized by GW6471, a PPARɑ inhibitor. Notably, erianin revealed vigoroso coordinate repression with LY294002 on AML cells. Our findings indicate that erianin showed a potent cytotoxic effect on AML cells and affected AML cells via PPARɑ to regulate PI3K/AKT signaling pathways. We demonstrated the potent anti-AML effects of erianin and reported its potential mechanisms of action, indicating its potential for further development as a novel anti-AML drug.
Collapse
Affiliation(s)
- Ying Deng
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Liang Zhong
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, China
| | - Yi Zhao
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Peng Wan
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Ying Zhang
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Yang Liao
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, China
| | - Hongyan Zhang
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Meng Wang
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China
| | - Beizhong Liu
- Clinical Laboratory of the Affiliated Yongchuan Hospital, Chongqing Medical University, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, China; Clinical Laboratory of the Affiliated Rehabilitation Hospital, Chongqing Medical University, China.
| |
Collapse
|
3
|
Kevat S, Mistry A, Oza N, Majmudar M, Patel N, Shah R, Ramachandran AV, Chauhan R, Haque S, Parashar NC, Tuli HS, Parashar G. Cancer Stem Cell Regulation as a Target of Therapeutic Intervention: Insights into Breast, Cervical and Lung Cancer. Cell Biochem Biophys 2025; 83:1521-1535. [PMID: 39843681 DOI: 10.1007/s12013-025-01666-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2025] [Indexed: 01/24/2025]
Abstract
Cancer Stem Cells (CSCs) play an important role in the development, resistance, and recurrence of many malignancies. These subpopulations of tumor cells have the potential to self-renew, differentiate, and resist conventional therapy, highlighting their importance in cancer etiology. This review explores the regulatory mechanisms of CSCs in breast, cervical, and lung cancers, highlighting their plasticity, self-renewal, and differentiation capabilities. CD44+/CD24- cells are a known marker for breast CSCs. Markers like as CD133 and ALDH have been discovered in cervical cancer CSCs. Similarly, in lung cancer, CSCs identified by CD44, CD133, and ALDH are linked to aggressive tumor behavior and poor therapy results. The commonalities between these tumors highlight the general necessity of targeting CSCs in treatment efforts. However, the intricacies of CSC activity, such as their interaction with the tumor microenvironment and particular signaling pathways differ between cancer types, demanding specialized methods. Wnt/β-catenin, Notch, and Hedgehog pathways are one of the essential signaling pathways, targeting them, may show ameliorative effects on breast, lung and cervical carcinomas and their respective CSCs. Pre-clinical data suggests targeting specific signaling pathways can eliminate CSCs, but ongoing clinical trials are on utilizing signaling pathway inhibitors in patients. In recent studies it has been reported that CAR T based targeting of specific markers may be used as combination therapy. Ongoing research related to nanobiotechnology can also play a significant role in diagnosis and treatment purpose targeting CSCs, as nanomaterials can be used for precise targeting and identification of CSCs. Further research into the targeting of signaling pathways and its precursors could prove to be right step into directing therapies towards CSCs for cancer therapy.
Collapse
Affiliation(s)
- Sakshi Kevat
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Archie Mistry
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Naman Oza
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Mohit Majmudar
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Netra Patel
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Rushabh Shah
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - A V Ramachandran
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- School Of Medicine, Universidad Espiritu Santo, Samborondon, Ecuador
| | | | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Gaurav Parashar
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India.
| |
Collapse
|
4
|
Hua W, Li F, Yang P, Lu Z, Liu Y, Zhong B, Shen B. Resveratrol derivative modified Ru(II) complexes: Synthesis, characterization, in vitro and in vivo anticancer study. J Inorg Biochem 2025; 267:112873. [PMID: 40048805 DOI: 10.1016/j.jinorgbio.2025.112873] [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: 12/02/2024] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/15/2025]
Abstract
The diversification of ligands provides more opportunities to adjust the photophysical performance as well as the bio-function of Ru(II) complexes as novel photosensitizers. Herein, a kind of Ru(II) complexes carrying resveratrol derivative, amino-Res, as ligand was designed and synthesized. The representative complex (named Ru4) showed potent anticancer activity under the trigger of 520 nm-light. Lipophilicity and cellular accumulation experiments indicated that Ru4 possessed higher LogPO/W value and cell up-take than Ru1-Ru3 and [Ru(bpy)3]2+. Mechanism study revealed that Ru4 could inhibit cancer cell migration, invasion and cancer stemness. The bio-function of Ru4 was mainly inherited from the amino-Res ligand. The in vivo study demonstrated that Ru4 could inhibit the tumor growth without significant system toxicity.
Collapse
Affiliation(s)
- Wuyang Hua
- School of Food Science and Nutrition Engineering, Jilin Agricultural Science and Technology University, 77(th) Han Lin Road, Jilin City 132101, China; Jilin Province Brewing Technology Science and Technology Innovation Center, 77(th) Han Lin Road, Jilin City 132101, China.
| | - Fenglin Li
- School of Food Science and Nutrition Engineering, Jilin Agricultural Science and Technology University, 77(th) Han Lin Road, Jilin City 132101, China; Jilin Province Brewing Technology Science and Technology Innovation Center, 77(th) Han Lin Road, Jilin City 132101, China
| | - Ping Yang
- School of Food Science and Nutrition Engineering, Jilin Agricultural Science and Technology University, 77(th) Han Lin Road, Jilin City 132101, China; Jilin Province Brewing Technology Science and Technology Innovation Center, 77(th) Han Lin Road, Jilin City 132101, China
| | - Zhongkui Lu
- School of Food Science and Nutrition Engineering, Jilin Agricultural Science and Technology University, 77(th) Han Lin Road, Jilin City 132101, China; Jilin Province Brewing Technology Science and Technology Innovation Center, 77(th) Han Lin Road, Jilin City 132101, China
| | - Yanxia Liu
- School of Food Science and Nutrition Engineering, Jilin Agricultural Science and Technology University, 77(th) Han Lin Road, Jilin City 132101, China; Jilin Province Brewing Technology Science and Technology Innovation Center, 77(th) Han Lin Road, Jilin City 132101, China
| | - Bao Zhong
- School of Food Science and Nutrition Engineering, Jilin Agricultural Science and Technology University, 77(th) Han Lin Road, Jilin City 132101, China; Jilin Province Brewing Technology Science and Technology Innovation Center, 77(th) Han Lin Road, Jilin City 132101, China
| | - Baoxing Shen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2(nd) Xue Lin Road, Nanjing 210023, China.
| |
Collapse
|
5
|
Chen Y, Qin Z, Wang Y, Gu B, Wang J, Zheng Y, Niu Y, Jia L. CD44-targeted virus-mimicking nanomedicine eliminates cancer stem cells and mitigates chemoresistance in head and neck squamous cell carcinoma. Mater Today Bio 2025; 32:101721. [PMID: 40242481 PMCID: PMC12002834 DOI: 10.1016/j.mtbio.2025.101721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/06/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) play critical roles in tumor growth, metastasis, and chemoresistance. Although several small-molecule inhibitors designed to inhibit CSCs have been investigated in clinical trials, their inadequate tumor targeting and potential off-target side effects have led to poor outcomes. A CD44-targeted virus-mimicking nanomedicine encapsulating the BMI1 inhibitor PTC209 (PTC209@VNP-HA) was designed to treat head and neck squamous cell carcinoma (HNSCC). We used a dendritic mesoporous silica nanoparticle (MSN) as the core for virus-mimicking nanoparticle (VNP) formation after adding shell particles to the MSN surface. The VNP surface was then modified with hyaluronic acid (HA), and PTC209 was adsorbed by mesopores to form PTC209@VNP-HA. In this system, HA is used to target CD44+ CSCs. The rough surface of VNP-HA provided better drug delivery efficiency than smooth nanoparticles modified with HA. VNP-HA enhanced the cancer inhibitory effect of PTC209 12-fold compared to the administration of free PTC209, leading to significantly higher bioavailability of PTC209. Both in vitro and in vivo assays showed that PTC209@VNP-HA inhibited cancer stemness, proliferation, and metastasis in HNSCC. Mechanistically, this inhibitory effect is closely associated with DNA damage/apoptosis signaling. Using a series of preclinical models in murine systems, we confirmed that PTC209@VNP-HA eliminated BMI1+ CSCs, and greatly inhibited the proliferation and metastasis of HNSCC when combined with cisplatin. This study investigated PTC209@VNP-HA as a novel and potentially transformative HNSCC treatment option that eliminates CSCs, prevents metastasis, and overcomes cisplatin resistance.
Collapse
Affiliation(s)
- Yiwen Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China
| | - Zhen Qin
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yujia Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Baoxin Gu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100091, PR China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yuting Niu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Lingfei Jia
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China
- Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, Peking University, Beijing 100091, PR China
| |
Collapse
|
6
|
Gao F, Liu S, Sun Y, Yu C, Zheng L, Sun L, Wang G, Sun Y, Bao Y, Song Z, Yang X, Ke C. Testes-specific protease 50 heightens stem-like properties and improves mitochondrial function in colorectal cancer. Life Sci 2025; 370:123560. [PMID: 40086746 DOI: 10.1016/j.lfs.2025.123560] [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: 10/29/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
AIMS The progression of colorectal cancer (CRC) is driven by a small subset of cancer stem-like cells (CSCs), and mitochondrial function is essential for maintaining their stemness. TSP50, a novel identified oncogene, has been found to promote cell proliferation in multiple cancer types. In this study, we detected the regulatory role of TSP50 in regulating CSC-like properties and mitochondrial mass in CRC. MATERIALS AND METHODS First, TSP50 expression and clinical relevance were analyzed via clinical databases and immunohistochemical (IHC). Subsequently, bioinformatic analyses, CRC cell lines, tumorsphere cultures, and mouse xenograft models were utilized to evaluate the relationship between TSP50 and CSC-like properties as well as mitochondrial mass. Finally, immunofluorescence, immunoprecipitation, and Western blotting were performed to dissect the regulatory mechanisms of TSP50, followed by rescue experiments conducted both in vitro and in vivo. KEY FINDINGS TSP50 was overexpressed in CRC tissues, correlating with poor drug response and shorter overall survival (OS). Meanwhile, TSP50 was shown to enhance CSC-like properties in both CRC cells and mouse xenograft models, while concurrently increasing mitochondrial mass and reducing ROS levels, these effects were partially reversed by inhibition of the PI3K/AKT pathway. Mechanistic investigations revealed that TSP50-induced activation of PI3K/AKT signaling is primarily mediated by the enhanced catalytic activity of PI3K p110α subunit. SIGNIFICANCE Collectively, TSP50 drives CRC malignancy by promoting CSC-like properties and enhancing mitochondrial function through PI3K/AKT signaling. These findings identify TSP50 as a potential therapeutic target for eliminating CSC-like cells and improving clinical outcomes in CRC treatment.
Collapse
Affiliation(s)
- Feng Gao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China; China International Joint Research Center for Human Stem Cell Bank, Northeast Normal University, Changchun, Jilin 130024, China
| | - Sichen Liu
- Division of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Laboratory of Surgical Translational Medicine, Jilin Provincial Engineering Laboratory for Thyroid Disease Control, Jilin, Changchun 130033, China; Department of Neurosurgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, China
| | - Yue Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Chunlei Yu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Lihua Zheng
- China International Joint Research Center for Human Stem Cell Bank, Northeast Normal University, Changchun, Jilin 130024, China
| | - Luguo Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Guannan Wang
- China International Joint Research Center for Human Stem Cell Bank, Northeast Normal University, Changchun, Jilin 130024, China
| | - Ying Sun
- China International Joint Research Center for Human Stem Cell Bank, Northeast Normal University, Changchun, Jilin 130024, China
| | - Yongli Bao
- China International Joint Research Center for Human Stem Cell Bank, Northeast Normal University, Changchun, Jilin 130024, China
| | - Zhenbo Song
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China
| | - Xiaoguang Yang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117, China.
| | - Chao Ke
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, China.
| |
Collapse
|
7
|
Manjili DA, Babaei FN, Younesirad T, Ghadir S, Askari H, Daraei A. Dysregulated circular RNA and long non-coding RNA-Mediated regulatory competing endogenous RNA networks (ceRNETs) in ovarian and cervical cancers: A non-coding RNA-Mediated mechanism of chemotherapeutic resistance with new emerging clinical capacities. Arch Biochem Biophys 2025; 768:110389. [PMID: 40090441 DOI: 10.1016/j.abb.2025.110389] [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: 11/11/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Cervical cancer (CC) and ovarian cancer (OC) are among the most common gynecological cancers with significant mortality in women, and their incidence is increasing. In addition to the prominent role of the malignant aspect of these cancers in cancer-related women deaths, chemotherapy drug resistance is a major factor that contributes to their mortality and presents a clinical obstacle. Although the exact mechanisms behind the chemoresistance in these cancers has not been revealed, accumulating evidence points to the dysregulation of non-coding RNAs (ncRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as key contributors. These ncRNAs perform the roles of regulators of signaling pathways linked to tumor formation and chemoresistance. Strong data from various recent studies have uncovered that the main mechanism of these ncRNAs in the induction of chemoresistance of CC and OC is done through a dysregulated miRNA sponge activity as competing endogenous RNA (ceRNA) in the competing endogenous RNA networks (ceRNETs), where a miRNA regulating a messenger RNA (mRNA) is trapped, thereby removing its inhibitory effect on the desired mRNA. Understanding these mechanisms is essential to enhancing treatment outcomes and managing the problem of drug resistance. This review provides a comprehensive overview of lncRNA- and circRNA-mediated ceRNETs as the core process of chemoresistance against the commonly used chemotherapeutics, including cisplatin, paclitaxel, oxaliplatin, carboplatin, and docetaxel in CC and OC. Furthermore, we highlight the clinical potential of these ncRNAs serving as diagnostic indicators of chemotherapy responses and therapeutic targets.
Collapse
Affiliation(s)
- Danial Amiri Manjili
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Naghdi Babaei
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tayebeh Younesirad
- Department of Medical Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sara Ghadir
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Askari
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
8
|
Berk Ş. Comprehensive bibliometric analysis and perspectives on therapies targeting colon cancer stem cells over a 40-year period. Regen Ther 2025; 29:19-34. [PMID: 40124468 PMCID: PMC11930536 DOI: 10.1016/j.reth.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
The presence of cancer stem cells (CSCs) is one of the primary causes of recurring therapy resistance because they have two main capacities: self-renewal and avoiding apoptotic pathways. Despite their relevance, no full bibliometric analysis has yet been done in this topic. The goal of this work is to use bibliometric analysis to map the fundamental and emergent areas in therapeutics targeting colon cancer stem cells. To perform bibliometric analysis on colon cancer stem cells (CCSCs) literature, spanning roughly the last 40 years, in order to establish a firm base for future projections by emphasizing the findings of the most notable research. All information pertinent to CCSCs was accessed from Web of Science Core Collection database. In order to identify and analyze the research hotspots and trends related to this topic, Biblioshiny (RStudio) and VOSviewer were utilized to ascertain the countries/regions, institutions, journals, authors, references, and keywords involved. The targeted time span covered 1735 research-, and review articles. The most frequent keywords were "colorectal cancer," "cancer stem cells," and "colon cancer," while the most trending keywords in the last few years were "protein stability," "spheroid formation," "ubiquitination," "exosomes," "patient-derived organoids," and "gut microbiota." Over the past 40 years, there has been a significant advancement in researchers' understanding of colon cancer stem cells. In addition, the cluster map of co-cited literature showed that colon cancer stem cell research has emerged as a research hotspot. It was also anticipated that the main focus of the future efforts appears to involve clinical applications of cell-targeted colon cancer therapy. These results provide researchers with a comprehensive understanding of this field and provide insightful ideas for further research.
Collapse
Affiliation(s)
- Şeyda Berk
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, 58140, Turkey
| |
Collapse
|
9
|
Subbiahanadar Chelladurai K, Selvan Christyraj JD, Rajagopalan K, Vadivelu K, Chandrasekar M, Das P, Kalimuthu K, Balamurugan N, Subramanian V, Selvan Christyraj JRS. Ex vivo functional whole organ in biomedical research: a review. J Artif Organs 2025; 28:131-145. [PMID: 39592544 DOI: 10.1007/s10047-024-01478-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/29/2024] [Indexed: 11/28/2024]
Abstract
Model systems are critical in biomedical and preclinical research. Animal and in vitro models serve an important role in our current understanding of human physiology, disease pathophysiology, and therapy development. However, if the system is between cell culture and animal models, it may be able to overcome the knowledge gap that exists in the current system. Studies employing ex vivo organs as models have not been thoroughly investigated. Though the integration of other organs and systems has an impact on many biological mechanisms and disorders, it can add a new dimension to modeling and aid in the identification of new possible therapeutic targets. Here, we have discussed why the ex vivo organ model is desirable and the importance of the inclusion of organs from diverse species, described its historical aspects, studied organs as models in scientific research, and its ex vivo stability. We also discussed, how an ex vivo organ model might help researchers better understand organ physiology, as well as organ-specific diseases and therapeutic targets. We emphasized how this ex vivo organ dynamics will be more competent than existing models, as well as what tissues or organs would have potentially viable longevity for ex vivo modeling including human tissues, organs, and/or at least biopsies and its possible advantage in clinical medicine including organ transplantation procedure and precision medicine.
Collapse
Affiliation(s)
- Karthikeyan Subbiahanadar Chelladurai
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Jackson Durairaj Selvan Christyraj
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India.
| | - Kamarajan Rajagopalan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Kayalvizhi Vadivelu
- Department of Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Meikandan Chandrasekar
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Puja Das
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Kalishwaralal Kalimuthu
- Rajiv Gandhi Centre for Biotechnology, Department of Biotechnology, Thiruvananthapuram, Kerala, India
| | - Nivedha Balamurugan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Vijayalakshmi Subramanian
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Johnson Retnaraj Samuel Selvan Christyraj
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India.
| |
Collapse
|
10
|
Chen S, Wang Y, Da Zhou, Hu J. Bayesian Inference of Phenotypic Plasticity of Cancer Cells Based on Dynamic Model for Temporal Cell Proportion Data. Biom J 2025; 67:e70055. [PMID: 40298362 DOI: 10.1002/bimj.70055] [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: 06/15/2024] [Revised: 09/18/2024] [Accepted: 10/31/2024] [Indexed: 04/30/2025]
Abstract
Mounting evidence underscores the prevalent hierarchical organization of cancer tissues. At the foundation of this hierarchy reside cancer stem cells, a subset of cells endowed with the pivotal role of engendering the entire cancer tissue through cell differentiation. In recent times, substantial attention has been directed toward the phenomenon of cancer cell plasticity, where the dynamic interconversion between cancer stem cells and nonstem cancer cells has garnered significant interest. Since the task of detecting cancer cell plasticity from empirical data remains a formidable challenge, we propose a Bayesian statistical framework designed to infer phenotypic plasticity within cancer cells, utilizing temporal data on cancer stem cell proportions. Our approach is grounded in a stochastic model, adept at capturing the dynamic behaviors of cells. Leveraging Bayesian analysis, we scrutinize the moment equation governing cancer stem cell proportions, derived from the Kolmogorov forward equation of our stochastic model. Our methodology introduces an improved Euler method for parameter estimation within nonlinear ordinary differential equation models, also extending insights to compositional data. Extensive simulations robustly validate the efficacy of our proposed method. To further corroborate our findings, we apply our approach to analyze published data from SW620 colon cancer cell lines. Our results harmonize with in situ experiments, thereby reinforcing the utility of our method in discerning and quantifying phenotypic plasticity within cancer cells.
Collapse
Affiliation(s)
- Shuli Chen
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong, China
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| | - Yuman Wang
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| | - Da Zhou
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| | - Jie Hu
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
11
|
Schachtschneider KM, Redlon LN, Lokken RP, Huang YH, Guzman G, Schook LB, Gaba RC. Epigenetic regulation of individual components of combined hepatocellular-cholangiocarcinoma. PLoS One 2025; 20:e0324145. [PMID: 40424447 PMCID: PMC12112136 DOI: 10.1371/journal.pone.0324145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Combined hepatocellular carcinoma-cholangiocarcinoma (HCC-CCA) is a rare liver tumor comprising histologic features of both HCC and CCA. Due to its heterogeneous nature, treatment of combined HCC-CCA is a significant clinical challenge and prognosis remains poor. Therefore, further understanding of the tumor biology underlying the individual subtypes of this mixed tumor is required to improve treatment stratification and optimize treatment strategies. This study sought to identify altered epigenetic regulation and gene expression patterns in the individual components of combined HCC-CCA. Formalin fixed paraffin embedded (FFPE) tumor specimens from 9 patients diagnosed with combined HCC-CCA were utilized in this study. Hematoxylin and eosin (H&E) staining was performed for each sample, and regions representative of the individual HCC and CCA components were delineated. Adjacent unstained slides were cut and dissected to separate HCC and CCA components. DNA and RNA extraction was performed for each sample for DNA methylation (n = 7 HCC and 7 CCA) and gene expression (n = 7 HCC and 8 CCA) profiling via reduced representation bisulfite sequencing (RRBS) and RNA-seq, respectively. Samples did not cluster by tumor type when comparing genome-wide DNA methylation or gene expression patterns. Of the 5 patients with DNA methylation data available for both subtypes, 4 clustered by patient as opposed to cancer subtype, suggesting similar epigenetic regulatory patterns arising from development in the same microenvironment and genetic background. Differential analysis resulted in the identification of 57 differentially expressed genes (DEGs) and 808 differentially methylated regions (DMRs) between the HCC and CCA subtypes. Genes associated with DMRs were associated with Wnt signaling, voltage-gated channels, metal binding, and cellular regulation. Finally, increased expression of several genes previously implicated in tumor aggressiveness, prognosis, and treatment responses were identified. These results highlight the potential importance of accounting for underlying HCC and CCA tumor biology when determining the optimal course of treatment for this deadly disease.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Luke N. Redlon
- College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ryan Peter Lokken
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, United States of America
| | - Yu-Hui Huang
- Department of Radiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Ron C. Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
12
|
Farhan M, Ling Z, Ding J, Shah Z, Dobrotae R. A novel fractional computational neural framework for analyzing cancer model under chemotherapy drug. Comput Methods Biomech Biomed Engin 2025:1-19. [PMID: 40418707 DOI: 10.1080/10255842.2025.2508227] [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: 01/25/2025] [Revised: 04/04/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025]
Abstract
In this study, a novel Caputo fractional-order model is proposed to represent the complex interactions among stem cells, effector cells, and tumor cells, considering both scenarios of chemotherapy. Furthermore, the proposed model, which incorporates treatment with effective chemotherapy, is thoroughly examined. The necessary properties, including the positivity and equilibrium points, as well as the local asymptotic stability analysis, are investigated. Additionally, the existence and uniqueness of solutions for the proposed model are thoroughly analyzed. We perform a thorough assessment of the solutions produced by the deep neural network by comparing them against established benchmarks and carefully analyzing them through testing, validation, training, error distribution analysis, and regression analysis. The temporal concentration pattern of stem, effector and tumor cells as well as chemotherapy drugs are examined. It is noted that chemotherapy leads to a decrease in tumor cell density over time, which extends the period required to achieve equilibrium. The decay rates of stem cells and tumor cells are recognized as essential elements affecting cancer dynamics. Furthermore, the integration of fractional orders is found to be important for precisely depicting the concentrations of cancer cells.
Collapse
Affiliation(s)
- Muhammad Farhan
- School of Mathematical Science, Yangzhou University, Yangzhou, China
| | - Zhi Ling
- School of Mathematical Science, Yangzhou University, Yangzhou, China
| | - Jie Ding
- School of Computer, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Zahir Shah
- Department of Mathematical Sciences, University of Lakki Marwat, Lakki Marwat, KPK, Pakistan
| | - Robert Dobrotae
- Faculty of Medicine, University of Medicine and Pharmacy 'Carol Davila', Bucharest, Romania
| |
Collapse
|
13
|
Xu K, Wu Q, Lingyun Z, Nguyen R, Safri F, Yang W, Xu Y, Ye Y, Kwan HY, Wang Q, Liang X, Shiddiky MJA, Warkiani ME, George J, Bao J, Qiao L. Extracellular vesicles as a promising platform of precision medicine in liver cancer. Pharmacol Res 2025:107800. [PMID: 40419123 DOI: 10.1016/j.phrs.2025.107800] [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: 02/19/2025] [Revised: 05/19/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025]
Abstract
Extracellular vesicles (EVs) are natural carriers of biological information and play pivotal roles in intercellular communication. EVs are biocompatible, have low immunogenicity, and are capable of traversing biological barriers, making them ideal tools for disease diagnosis and therapy. Despite their promising prospects, the full realization of EVs potential faces several challenges. This article aims to comprehensively review the biological and molecular features of EVs, their applications in liver cancer and possible underlying mechanisms, and the critical challenges affecting the clinical translation of EVs-based therapies in liver cancer.
Collapse
Affiliation(s)
- Keyang Xu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Zhao Lingyun
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Romario Nguyen
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Fatema Safri
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - William Yang
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Yikun Xu
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Yun Ye
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong, China
| | - Hiu Yee Kwan
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong, China
| | - Qiang Wang
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Xiuming Liang
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine Karolinska Institute, Stockholm, Sweden
| | - Muhammad J A Shiddiky
- Rural Health Research Institute (RHRI), Charles Sturt University, Orange NSW 2800, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, the University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Jianfeng Bao
- Hangzhou Xixi Hospital affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia.
| |
Collapse
|
14
|
Xie G, Okuda S, Gao JY, Wu T, Jeong J, Lu KP, Zhou XZ. The Central Role of Pin1 in Age-Related Cancer Signaling Pathways. Semin Cancer Biol 2025:S1044-579X(25)00072-0. [PMID: 40412492 DOI: 10.1016/j.semcancer.2025.05.011] [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: 01/17/2025] [Revised: 04/05/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
The prolyl-isomerase Pin1 is a unique enzyme that catalyzes cis-trans isomerization of phosphorylated Ser/Thr-Pro motifs. These motifs are present in many proteins, where isomerization of the typically rigid prolyl-peptide bond can lead to conformational changes, and subsequently regulate activity, stability, or localization. The specificity of Pin1 for phosphorylated motifs allows it to serve as a master regulator of proteins after phosphorylation, adding an additional layer of regulation to intricately control cellular signaling. As such, Pin1 plays an expansive role in numerous cancer and age-related signaling pathways, and is recognized as a major driver of cancer and promising therapeutic target. In this review, we discuss the role of Pin1 in regulation of age-related cancer signaling pathways, and we highlight the early development and current landscape of Pin1 inhibitors, and the prospect of Pin1 inhibition for cancer therapy.
Collapse
Affiliation(s)
- George Xie
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Sho Okuda
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Jing-Yan Gao
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Department of Chemistry, Western University, London, ON N6A 5C1, Canada
| | - Timothy Wu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Jessica Jeong
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON N6G 2V4, Canada.
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada; Lawson Health Research Institute, Western University, London, ON N6C 2R5, Canada.
| |
Collapse
|
15
|
Yamaguchi H, Okada M, Otani T, On J, Shibuma S, Takino T, Watanabe J, Tsukamoto Y, Ogura R, Oishi M, Suzuki T, Ishikawa A, Sakata H, Natsumeda M. Near-Infrared Photoimmunotherapy in Brain Tumors-An Unexplored Frontier. Pharmaceuticals (Basel) 2025; 18:751. [PMID: 40430568 PMCID: PMC12115099 DOI: 10.3390/ph18050751] [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: 03/31/2025] [Revised: 04/16/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a promising cancer treatment that uses near-infrared light to activate a conjugate of a monoclonal antibody (mAb) and a photoactivatable silica phthalocyanine dye (IRDye700DX: IR700). Unlike conventional photodynamic therapy (PDT), NIR-PIT selectively destroys targeted tumor cells while preserving the surrounding normal tissue and providing superior tissue penetration. Recently, NIR-PIT has been approved for the treatment of unresectable recurrent head and neck cancers in Japan. It induces highly selective cancer cell death; therefore, it is expected to be a new curative treatment option for various cancers, including brain tumors. In this review, we compare the principles of NIR-PIT and PDT and discuss the potential applications of NIR-PIT for brain tumors. We selected targetable proteins across various types of brain tumors and devised a strategy to effectively pass the mAb-IR700 conjugate through the blood-brain barrier (BBB), which is a significant challenge for NIR-PIT in treating brain tumors. Innovative approaches for delivering the mAb-IR700 conjugate across the BBB include exosomes, nanoparticle-based systems, and cell-penetrating peptides. Small-molecule compounds, such as affibodies, are anticipated to rapidly accumulate in tumors within intracranial models, and our preliminary experiments demonstrated rapid uptake. NIR-PIT also induces immunogenic cell death and activates the anti-tumor immune response. Overall, NIR-PIT is a promising approach for treating brain tumors. It has the potential to overcome the limitations of conventional therapies and offers new hope to patients with brain tumors.
Collapse
Affiliation(s)
- Haruka Yamaguchi
- Department of Biochemistry, School of Life Dentistry at Niigata, The Nippon Dental University, Niigata 951-8580, Japan;
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Takuya Otani
- Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan;
| | - Jotaro On
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
| | - Satoshi Shibuma
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
| | - Toru Takino
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
| | - Jun Watanabe
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
| | - Ryosuke Ogura
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
| | - Makoto Oishi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
| | - Takamasa Suzuki
- Faculty of Engineering, Niigata University, Niigata 950-2181, Japan;
| | - Akihiro Ishikawa
- Startup Incubation Center, Shimadzu Corporation, Kyoto 604-8511, Japan; (A.I.); (H.S.)
| | - Hideyuki Sakata
- Startup Incubation Center, Shimadzu Corporation, Kyoto 604-8511, Japan; (A.I.); (H.S.)
| | - Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.O.); (J.O.); (S.S.); (T.T.); (J.W.); (Y.T.); (R.O.); (M.O.)
- Advanced Treatment of Neurological Diseases Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| |
Collapse
|
16
|
Das A, Yilmaz O, Yilmaz O, Deshpande V. SOX17: a new therapeutic target for immune evasion of colorectal cancer. J Clin Pathol 2025:jcp-2024-209878. [PMID: 40350244 DOI: 10.1136/jcp-2024-209878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 05/14/2025]
Abstract
Despite advances in cancer immunotherapies across various cancers, survival outcomes in colorectal cancer (CRC) with these agents remain largely unsatisfactory despite the high tumour burden. Colorectal stem cells (CSCs), especially LGR5+ CSCs, are the significant drivers in CRC initiation, progression and resistance to conventional therapies. Although native immune surveillance is sufficient to combat early tumour formation, CRC evades early immune detection with its well-documented adenoma-to-carcinoma sequence. The exact mechanism underlying this phenomenon still needs to be better understood. SRY-related HMG box gene 17 (SOX17), a transcription factor that specifies embryonic gut formation, is increasingly recognised as a significant factor in CRC tumourigenesis. However, its role as a tumour suppressor or oncogene is still debated. Evidence from a recent study highlighted the critical role of SOX17 in reshaping the tumour immune ecosystem through the simultaneous inhibition of CD8+ T cells and selective suppression of LGR5 expression in CSCs through transcriptional repression, thereby facilitating disease progression. Given its role in immune evasion, SOX17 could be a promising marker in personalised therapy. Additionally, SOX17 could play a role in the diagnostic arena, potentially identifying dysplasia in the gastrointestinal tract. Future clinical, basic and genetic studies focusing on SOX17 are needed to ascertain its mechanistic role in tumour immunomodulation in CRC and diagnosing preneoplastic lesions in the gastrointestinal tract.
Collapse
Affiliation(s)
- Avash Das
- Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Omer Yilmaz
- Harvard Medical School, Boston, Massachusetts, USA
| | - Osman Yilmaz
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Vikram Deshpande
- Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Zike AB, Abel MG, Fleck SA, DeWitt ED, Weaver LN. Estrogen-related receptor is required in adult Drosophila females for germline stem cell maintenance. Dev Biol 2025; 524:132-143. [PMID: 40348318 DOI: 10.1016/j.ydbio.2025.05.005] [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: 02/10/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Adult tissue function is dependent on intrinsic factors that mediate stem cell self-renewal and proliferation in response to changes in physiology and the environment. The estrogen-related receptor (ERR) subfamily of orphan nuclear receptors are major transcriptional regulators of metabolism and animal physiology. In mammals, ERRs (NR3B1, NR3B2, NR3B3) have roles in regulating mitochondrial biosynthesis, lipid metabolism, as well as stem cell maintenance. The sole Drosophila ERR ortholog promotes larval growth by establishing a metabolic state during the latter half of embryogenesis. In addition, ERR is required in adult Drosophila males to coordinate glycolytic metabolism with lipid synthesis and within the testis to regulate spermatogenesis gene expression and fertility. Despite extensive work characterizing the role of ERR in Drosophila metabolism, whether ERR has a conserved requirement in regulating stem cell behavior has been understudied. To determine whether ERR regulates stem cell activity in Drosophila, we used the established adult female germline stem cell (GSC) lineage as a model. We found that whole-body ERR knockout in adult females using conditional heat shock-driven FLP-FRT recombination significantly decreases GSC number and glycolytic enzyme expression in GSCs. In addition, we found that ERR activity is required cell-autonomously in the adult female germline for maintenance of GSCs; whereas ERR regulation of GSCs is independent of its activity in adult female adipocytes. Our results highlight an ancient and conserved role for ERRs in the regulation of stem cell self-renewal.
Collapse
Affiliation(s)
- Anna B Zike
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Madison G Abel
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Sophie A Fleck
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Emily D DeWitt
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Lesley N Weaver
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
18
|
Room SA, Chen PJ, Chen ZY, Shih YJ, Pan SY, Hsu YC, Hsiao TC, Ting YC, Chou CCK, Wu CH, Chi KH. Chemical characterization and oxidative potential of persistent organic pollutants (POPs) in size-resolved particulate matter across industrial and traffic stations. ENVIRONMENTAL RESEARCH 2025; 279:121747. [PMID: 40320031 DOI: 10.1016/j.envres.2025.121747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/15/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
This study is the first to investigate polychlorinated dibenzo-p-dioxins and dibenzofurans (PCDD/Fs), polychlorinated biphenyls (PCBs), and polychlorinated naphthalenes (PCNs) across multiple particulate matter (PM) sizes (PM1.0, PM2.5, TSP) in Taiwan, focusing on spatio-seasonal variations, chemical composition, sources, and oxidative potential (OP) utilizing Real-time Cell Analysis (RTCA) and the Dithiothreitol (DTT) assay. PM samples were collected from the Northern Industrial Station (NIS: PM) in Taoyuan, and the Central Industrial (CIS: PM2.5) and Central Traffic (CTS: PM2.5) stations in Taichung (2022-2023). Elevated PCDD/F, PCB, and PCN levels were observed at NIS during winter, with PM2.5 and PM1.0 comprising 90 % and 50 % of TSP, respectively, driven by local emissions and meteorological influences. PCDD/Fs peaked in winter at CTS (7.16 ± 1.64 fg TEQWHO/m3) and in autumn at CIS (8.29 ± 3.21 fg TEQWHO/m3), while PCBs were highest in summer (CIS: 0.151 ± 0.212 fg TEQWHO/m3; CTS: 0.006 ± 0.013 fg TEQWHO/m3), likely due to temperature-driven volatilization. Notably, PCNs exhibited no clear seasonal trends. Cytotoxicity assays revealed a size-dependent toxicity gradient (PM1.0: 71.8 % > PM2.5: 62.1 % > TSP: 51.9 %), with PM2.5 toxicity consistent across sources (P = 0.58). DTT assays indicated higher OP at Northern Taiwan's industrial site on weekdays, whereas Central Taiwan's industrial and traffic sites showed no substantial variation (p > 0.05). Markedly, NO3- strongly correlated with OP across all PM sizes, while Cu and Cr were linked to OPv, and Mn and Cr to OPm. These findings highlight seasonal and source-driven PM toxicity, with smaller particles posing greater health risks, requiring targeted mitigation.
Collapse
Affiliation(s)
- Shahzada Amani Room
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Po Jui Chen
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Zhi Yu Chen
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yu Ju Shih
- Department of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Education, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Shih Yu Pan
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yuan-Cheng Hsu
- National Environmental Research Academy, Ministry of Environment, Taoyuan, 330, Taiwan
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taiwan
| | - Yu Chieh Ting
- Graduate Institute of Environmental Engineering, National Taiwan University, Taiwan
| | - Charless C-K Chou
- Research Center for Environmental Changes, Academia Sinica, Taipei, 115, Taiwan
| | - Chien-Hou Wu
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kai Hsien Chi
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
19
|
Guo Q, Qin H, Chen Z, Zhang W, Zheng L, Qin T. Key roles of ubiquitination in regulating critical regulators of cancer stem cell functionality. Genes Dis 2025; 12:101311. [PMID: 40034124 PMCID: PMC11875185 DOI: 10.1016/j.gendis.2024.101311] [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: 11/02/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 03/05/2025] Open
Abstract
The ubiquitin (Ub) system, a ubiquitous presence across eukaryotes, plays a crucial role in the precise orchestration of diverse cellular protein processes. From steering cellular signaling pathways and orchestrating cell cycle progression to guiding receptor trafficking and modulating immune responses, this process plays a crucial role in regulating various biological functions. The dysregulation of Ub-mediated signaling pathways in prevalent cancers ushers in a spectrum of clinical outcomes ranging from tumorigenesis and metastasis to recurrence and drug resistance. Ubiquitination, a linchpin process mediated by Ub, assumes a central mantle in molding cellular signaling dynamics. It navigates transitions in biological cues and ultimately shapes the destiny of proteins. Recent years have witnessed an upsurge in the momentum surrounding the development of protein-based therapeutics aimed at targeting the Ub system under the sway of cancer stem cells. The article provides a comprehensive overview of the ongoing in-depth discussions regarding the regulation of the Ub system and its impact on the development of cancer stem cells. Amidst the tapestry of insights, the article delves into the expansive roles of E3 Ub ligases, deubiquitinases, and transcription factors entwined with cancer stem cells. Furthermore, the spotlight turns to the interplay with pivotal signaling pathways the Notch, Hedgehog, Wnt/β-catenin, and Hippo-YAP signaling pathways all play crucial roles in the regulation of cancer stem cells followed by the specific modulation of Ub-proteasome.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou 550014, China
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Artificial Intelligence and IoT Smart Medical Engineering Research Center of Henan Province, Zhengzhou, Henan 450008, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Tingting Qin
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| |
Collapse
|
20
|
Wang Z, Zhang J, Luo L, Zhang C, Huang X, Liu S, Chen H, Miao W. Nucleoporin 93 Regulates Cancer Cell Growth and Stemness in Bladder Cancer via Wnt/β-Catenin Signaling. Mol Biotechnol 2025; 67:2072-2084. [PMID: 38744786 DOI: 10.1007/s12033-024-01184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024]
Abstract
Bladder cancer (BLCA) is a prevalent cancer type with an unmet need for new therapeutic strategies. Nucleoporin 93 (Nup93) is implicated in the pathophysiology of several cancers, but its relationship with bladder cancer remains unclear. Nup93 expression was analyzed in TCGA datasets and 88 BLCA patient samples. Survival analysis and Cox regression models evaluated the association between Nup93 levels and patient prognosis. BLCA cells were used to investigate the effects of Nup93 overexpression or knockdown on cell growth, invasion, stemness (sphere formation and ALDH2 + cancer stem cell marker), and Wnt/β-catenin signaling in vitro. The Wnt activator BML-284 was used to confirm the involvement of Wnt/β-catenin signaling pathway. A xenograft mouse model validated the in vitro findings. Nup93 was highly expressed in BLCA tissues and cell lines, and high Nup93 expression correlated with poor prognosis in BLCA patients. Nup93 silencing inhibited BLCA cell proliferation, Wnt/β-catenin activation, and cancer cell stemness. Conversely, Nup93 overexpression promoted these effects. BML-284 partially rescued the reduction in cell growth and stemness markers caused by Nup93 knockdown. Nup93 knockdown also suppressed the tumor formation of BLCA cells in vivo. Nup93 regulates BLCA cell growth and stemness via the Wnt/β-catenin pathway, suggesting its potential as a prognostic marker and therapeutic target in BLCA.
Collapse
Affiliation(s)
- Zhe Wang
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Jing Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Lina Luo
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Chao Zhang
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Xiaomeng Huang
- Medical Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Shuo Liu
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Huaian Chen
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Wenlong Miao
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China.
| |
Collapse
|
21
|
Qi H, Zhao H, Li E, Lu X, Yu N, Liu J, Han J. DeepQA: A Unified Transcriptome-Based Aging Clock Using Deep Neural Networks. Aging Cell 2025; 24:e14471. [PMID: 39757434 PMCID: PMC12074024 DOI: 10.1111/acel.14471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/21/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025] Open
Abstract
Understanding the complex biological process of aging is of great value, especially as it can help develop therapeutics to prolong healthy life. Predicting biological age from gene expression data has shown to be an effective means to quantify aging of a subject, and to identify molecular and cellular biomarkers of aging. A typical approach for estimating biological age, adopted by almost all existing aging clocks, is to train machine learning models only on healthy subjects, but to infer on both healthy and unhealthy subjects. However, the inherent bias in this approach results in inaccurate biological age as shown in this study. Moreover, almost all existing transcriptome-based aging clocks were built around an inefficient procedure of gene selection followed by conventional machine learning models such as elastic nets, linear discriminant analysis etc. To address these limitations, we proposed DeepQA, a unified aging clock based on mixture of experts. Unlike existing methods, DeepQA is equipped with a specially designed Hinge-Mean-Absolute-Error (Hinge-MAE) loss so that it can train on both healthy and unhealthy subjects of multiple cohorts to reduce the bias of inferring biological age of unhealthy subjects. Our experiments showed that DeepQA significantly outperformed existing methods for biological age estimation on both healthy and unhealthy subjects. In addition, our method avoids the inefficient exhaustive search of genes, and provides a novel means to identify genes activated in aging prediction, alternative to such as differential gene expression analysis.
Collapse
Affiliation(s)
- Hongqian Qi
- State Key Laboratory of Medicinal Chemical BiologyNankai UniversityTianjinChina
- College of PharmacyNankai UniversityTianjinChina
| | - Hongchen Zhao
- College of Artificial IntelligenceNankai UniversityTianjinChina
| | - Enyi Li
- College of Artificial IntelligenceNankai UniversityTianjinChina
| | - Xinyi Lu
- State Key Laboratory of Medicinal Chemical BiologyNankai UniversityTianjinChina
| | - Ningbo Yu
- College of Artificial IntelligenceNankai UniversityTianjinChina
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of EducationNankai UniversityChina
| | - Jinchao Liu
- College of Artificial IntelligenceNankai UniversityTianjinChina
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of EducationNankai UniversityChina
| | - Jianda Han
- College of Artificial IntelligenceNankai UniversityTianjinChina
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of EducationNankai UniversityChina
| |
Collapse
|
22
|
Higuchi Y, Teo JL, Yi D, Kahn M. Safely Targeting Cancer, the Wound That Never Heals, Utilizing CBP/Beta-Catenin Antagonists. Cancers (Basel) 2025; 17:1503. [PMID: 40361430 PMCID: PMC12071182 DOI: 10.3390/cancers17091503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Stem cells, both normal somatic (SSC) and cancer stem cells (CSC) exist in minimally two states, i.e., quiescent and activated. Regulation of these two states, including their reliance on different metabolic processes, i.e., FAO and glycolysis in quiescent versus activated stem cells respectively, involves the analysis of a complex array of factors (nutrient and oxygen levels, adhesion molecules, cytokines, etc.) to initiate the epigenetic changes to either depart or enter quiescence. Quiescence is a critical feature of SSC that is required to maintain the genomic integrity of the stem cell pool, particularly in long lived complex organisms. Quiescence in CSC, whether they are derived from mutations arising in SSC, aberrant microenvironmental regulation, or via dedifferentiation of more committed progenitors, is a critical component of therapy resistance and disease latency and relapse. At the beginning of vertebrate evolution, approximately 450 million years ago, a gene duplication generated the two members of the Kat3 family, CREBBP (CBP) and EP300 (p300). Despite their very high degree of homology, these two Kat3 coactivators play critical and non-redundant roles at enhancers and super-enhancers via acetylation of H3K27, thereby controlling stem cell quiescence versus activation and the cells metabolic requirements. In this review/perspective, we discuss the unique regulatory roles of CBP and p300 and how specifically targeting the CBP/β-catenin interaction utilizing small molecule antagonists, can correct lineage infidelity and safely eliminate quiescent CSC.
Collapse
Affiliation(s)
- Yusuke Higuchi
- Beckman Research Institute, City of Hope, Duarte, CA 91010, USA;
| | - Jia-Ling Teo
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (J.-L.T.); (D.Y.)
| | - Daniel Yi
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (J.-L.T.); (D.Y.)
| | - Michael Kahn
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (J.-L.T.); (D.Y.)
| |
Collapse
|
23
|
Rajan SS, Merlin JPJ, Abrahamse H. Breaking the Resistance: Photodynamic Therapy in Cancer Stem Cell-Driven Tumorigenesis. Pharmaceutics 2025; 17:559. [PMID: 40430852 PMCID: PMC12115314 DOI: 10.3390/pharmaceutics17050559] [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: 03/26/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Cancer stem cells (CSCs) are essential for the growth of malignancies because they encourage resistance to cancer therapy and make metastasis and relapse easier. To effectively tackle the obstacles presented by CSCs, novel therapeutic approaches are required. Photodynamic therapy (PDT) is a promising treatment option for cancer cells, which uses light-sensitive medications that are activated by light wavelengths. This review investigates the use of PDT to overcome malignancies driven by CSCs that have innate resistance mechanisms. PDT works by causing tumor cells to accumulate photosensitizers (PSs) selectively. The reactive oxygen species (ROS), which kill cells, are released by these PSs when they are stimulated by light. According to recent developments in PDT, its efficacy may go beyond traditional tumor cells, providing a viable remedy for the resistance shown by CSCs. Researchers want to improve the targeted elimination and selective targeting of CSCs by combining PDT with new PSs and customized delivery systems. Studies emphasize how PDT affects CSCs as well as bulk tumor cells. According to studies, PDT not only limits CSC growth but also modifies their microenvironment, which lowers the possibility of recovery. Additionally, studies are being conducted on the utilization of PDT and immunotherapeutic techniques to improve treatment efficacy and overcome inherent resistance of CSCs. In conclusion, PDT is a viable strategy for treating carcinogenesis driven by CSCs. By applying the most recent advancements in PDT technologies and recognizing how it interacts with CSCs, this treatment has the potential to surpass traditional resistance mechanisms and improve the future of cancer patients. Clinical and preclinical studies highlight that combining PDT with CSC-targeted approaches has the potential to overcome current therapy limitations. Future efforts should focus on clinical validation, optimizing light delivery and PS use, and developing effective combination strategies to target CSCs.
Collapse
Affiliation(s)
- Sheeja S. Rajan
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (J.P.J.M.); (H.A.)
| | | | | |
Collapse
|
24
|
Tamori S, Matsuda C, Kasai T, Ohno S, Sasaki K, Akimoto K. Asymmetric cell division of ALDH1-positive cancer stem cells generates glycolytic metabolically diverse cell populations. Sci Rep 2025; 15:13932. [PMID: 40263471 PMCID: PMC12015440 DOI: 10.1038/s41598-025-97985-2] [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: 08/28/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
Metabolic heterogeneity in various cancer cells within a tumor causes resistance to medical therapies and promotes tumor recurrence and metastasis. However, the mechanisms by which tumors acquire metabolic heterogeneity are poorly understood. Here, we revealed that PKCλ-dependent asymmetric division of ALDH1-positive cancer stem cells (CSCs) led to an uneven distribution of glycolytic capacity, which is crucial for understanding metabolic heterogeneity within a tumor. The rate-limiting enzyme PFKP and the metabolic probe CDG in glycolysis codistributed with the ALDH1A3 protein during the post-cell division phase, highlighting a mechanism for acquiring metabolic diversity. PKCλ deficiency reduced the asymmetric distribution of these proteins in ALDH1high cells with high ALDH1 activity, suggesting a fundamental role for PKCλ in metabolic heterogeneity. We identified 28 distinct distribution patterns combining PFKP and CDG distributions, demonstrating the complexity of glycolytic heterogeneity. Furthermore, validation and prediction of cell distribution patterns via a probabilistic model confirmed that PKCλ deficiency diminished glycolytic diversity in individual cells within a cancer cell colony generated from an ALDH1-positive CSC. These findings suggest that PKCλ-dependent asymmetric cell division of ALDH1-positive CSCs is crucial for glycolytic heterogeneity in cancer cells within a tumor, potentially offering new therapeutic targets against tumor resistance and metastasis.
Collapse
Affiliation(s)
- Shoma Tamori
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
- Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Chika Matsuda
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Takahiro Kasai
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Shigeo Ohno
- Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazunori Sasaki
- Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, Japan.
| | - Kazunori Akimoto
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan.
- Research Division of Medical Data Science, Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan.
| |
Collapse
|
25
|
Pagliari F, Tirinato L, Di Fabrizio E. Raman spectroscopies for cancer research and clinical applications: a focus on cancer stem cells. Stem Cells 2025; 43:sxae084. [PMID: 39949042 DOI: 10.1093/stmcls/sxae084] [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: 05/10/2024] [Accepted: 11/20/2024] [Indexed: 04/23/2025]
Abstract
Over the last 2 decades, research has increasingly focused on cancer stem cells (CSCs), considered responsible for tumor formation, resistance to therapies, and relapse. The traditional "static" CSC model used to describe tumor heterogeneity has been challenged by the evidence of CSC dynamic nature and plasticity. A comprehensive understanding of the mechanisms underlying this plasticity, and the capacity to unambiguously identify cancer markers to precisely target CSCs are crucial aspects for advancing cancer research and introducing more effective treatment strategies. In this context, Raman spectroscopy (RS) and specific Raman schemes, including CARS, SRS, SERS, have emerged as innovative tools for molecular analyses both in vitro and in vivo. In fact, these techniques have demonstrated considerable potential in the field of cancer detection, as well as in intraoperative settings, thanks to their label-free nature and minimal invasiveness. However, the RS integration in pre-clinical and clinical applications, particularly in the CSC field, remains limited. This review provides a concise overview of the historical development of RS and its advantages. Then, after introducing the CSC features and the challenges in targeting them with traditional methods, we review and discuss the current literature about the application of RS for revealing and characterizing CSCs and their inherent plasticity, including a brief paragraph about the integration of artificial intelligence with RS. By providing the possibility to better characterize the cellular diversity in their microenvironment, RS could revolutionize current diagnostic and therapeutic approaches, enabling early identification of CSCs and facilitating the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Francesca Pagliari
- Division of Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Luca Tirinato
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Enzo Di Fabrizio
- PolitoBIOMed Lab DISAT Department, Polytechnic University of Turin, 10129 Turin, Italy
| |
Collapse
|
26
|
Bardhan M, Muneer MA, Khare A, Minesh Shah R, Kaur A, Vasipalli SS, Suresh V, Podder V, Ahluwalia M, Odia Y, Chen Z. Advances in stem cell-based therapeutic transfers for glioblastoma treatment. Expert Rev Neurother 2025:1-17. [PMID: 40245098 DOI: 10.1080/14737175.2025.2490543] [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: 09/23/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Glioblastoma (GBM), a highly malignant brain tumor, has a poor prognosis despite standard treatments like surgery, chemotherapy, and radiation. Glioblastoma stem cells (GSCs) play a critical role in recurrence and therapy resistance. Stem cell-based therapies have emerged as innovative approaches, leveraging the tumor-targeting abilities of stem cells to deliver treatments directly to GBM. AREAS COVERED This review focuses on using intact stem cells or subtypes for GBM therapy, excluding antigenic characteristics. The stem cell-based therapies explored include neural, mesenchymal, glioblastoma, hematopoietic and adipose-derived stem cells that have been investigated in both clinical and preclinical settings. A systematic search in PubMed, EMBASE, ClinicalTrials.gov, and Scopus had identified research up until January 2024. Key mechanisms reviewed include immune modulation, angiogenesis inhibition, and apoptosis induction. Discussion of completed and ongoing trials include emphasis on safety, efficacy, challenges, and study design limitations. EXPERT OPINION Stem cell-based therapies hold promise for treating GBM by targeting GSCs and improving treatment outcomes. Despite some potential advantages, challenges such as tumorigenesis risks, delivery complexities, and sustained therapeutic effects persist. Future research should prioritize optimizing stem cell modifications, combining them with current treatments, and conducting large-scale trials to ensure safety and efficacy. Integrating stem cell therapies into GBM treatment could provide more effective and less invasive options for patients.
Collapse
Affiliation(s)
- Mainak Bardhan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | | - Abhinav Khare
- All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | | | - Anmol Kaur
- Lady Hardinge Medical College, New Delhi, India
| | - Sonit Sai Vasipalli
- Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Vinay Suresh
- King George's Medical University, Lucknow, India
| | - Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Manmeet Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Yazmin Odia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Zhijian Chen
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| |
Collapse
|
27
|
Souto EP, Gong P, Landua JD, Rajaram Srinivasan R, Ganesan A, Dobrolecki LE, Purdy SC, Pan X, Zeosky M, Chung A, Yi SS, Ford HL, Lewis MT. Lineage Tracing and Single-Cell RNA Sequencing Reveal a Common Transcriptional State in Breast Cancer Tumor-Initiating Cells Characterized by IFN/STAT1 Activity. Cancer Res 2025; 85:1390-1409. [PMID: 40230213 PMCID: PMC11997551 DOI: 10.1158/0008-5472.can-23-4022] [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: 12/19/2023] [Revised: 09/03/2024] [Accepted: 01/31/2025] [Indexed: 04/16/2025]
Abstract
A tumor cell subpopulation of tumor-initiating cells (TIC) or "cancer stem cells" is associated with therapeutic resistance, as well as both local and distant recurrences. Signal transducer and activator of transcription (STAT) activity is elevated in TICs in claudin-low models of human triple-negative breast cancer, which enables enrichment of TICs using a STAT-responsive reporter. Lineage tracing of TICs as they undergo cell state changes could enable a better understanding of the molecular phenotypes of TIC and uncover strategies to selectively target TICs. In this study, we developed a STAT-responsive lineage-tracing system and used it in conjunction with the original reporter to enrich for cells with enhanced mammosphere-forming potential. This approach was able to detect TICs in some, but not all, basal-like triple-negative breast cancer xenograft models, indicating that STAT signaling has both TIC-related and TIC-independent functions. Single-cell RNA sequencing (RNA-seq) of reporter-tagged xenografts and clinical samples identified a common IFN/STAT1-associated transcriptional state in TICs that was previously linked to inflammation and macrophage differentiation. Surprisingly, most of the identified genes were not present in previously published TIC signatures derived using bulk RNA-seq. Finally, bone marrow stromal cell antigen-2 was identified as a cell surface marker of this state that functionally regulated TIC frequency. These results suggest that TICs may exploit the IFN/STAT1 signaling axis to promote their activity and that targeting this pathway may help eliminate TICs. Significance: Coupling single-cell transcriptomics with tumor-initiating cell enrichment identified IFN response gene expression not previously reported in bulk RNA-sequencing-derived signatures and proposed IFN/STAT1 signaling as a candidate therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Eric P. Souto
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Ping Gong
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - John D. Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | | | - Abhinaya Ganesan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Stephen C. Purdy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Xingxin Pan
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Michael Zeosky
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Anna Chung
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - S. Stephen Yi
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
28
|
Chen J, Li H, Jin Q, Li X, Zhang Y, Shen J, Huang G, Yin J, Zou C, Li X, He X, Xie X, Lin T. Troxerutin suppresses the stemness of osteosarcoma via the CD155/SRC/β-catenin signaling axis. Cell Mol Biol Lett 2025; 30:45. [PMID: 40217455 PMCID: PMC11992710 DOI: 10.1186/s11658-025-00724-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Osteosarcoma is the most prevalent primary malignant bone tumor affecting pediatric and adolescent individuals. However, despite the passage of three decades, there has been no notable enhancement in the overall survival rate of patients with osteosarcoma. In recent years, CD155 has been reported to exhibit abnormal amplification in a range of tumors, yet the precise underlying mechanism remains elusive. The objective of this study is to investigate the role of CD155 in osteosarcoma, and to identify drugs that specifically target this molecule, thereby offering a novel direction for the treatment of osteosarcoma. METHODS The prognosis of patients with osteosarcoma with high and low expression of CD155 was verified by immunohistochemistry. CCK-8 and colony formation assays were used to detect cell proliferation and drug resistance. Transwell experiments were used to detect cell migration and invasion. The sphere formation experiment was used to evaluate the stemness of tumor cells. Additionally, in vivo animal models were utilized to assess the functional role of CD155 in a biological context. RNA-seq and co-immunoprecipitation methods were used to search for downstream target molecules and signaling pathways of CD155. Finally, virtual screening was used to find drugs targeting CD155. RESULTS In this study, we have established the significant amplification of CD155 in osteosarcoma. Utilizing a comprehensive array of experimental methods, including CCK-8 assay, colony formation assay, Transwell assay, and in vivo animal models, we unequivocally demonstrate that CD155 significantly potentiates the malignancy of osteosarcoma both in vitro and in vivo. Additionally, our findings reveal that CD155 promotes osteosarcoma stemness by modulating the Wnt/β-catenin signaling pathway. Advanced molecular techniques, such as RNA sequencing and co-immunoprecipitation, have been instrumental in elucidating the mechanism of CD155 in activating the Wnt/β-catenin pathway via the SRC/AKT/GSK3β signaling axis, thereby enhancing the stem-cell-like properties of osteosarcoma cells. To explore targeted therapeutic options, we conducted virtual screening and identified troxerutin as a promising CD155 inhibitor. CONCLUSIONS Our findings reveal that troxerutin effectively inhibits CD155, attenuates the SRC/AKT/GSK3β signaling cascade, diminishes the nuclear localization of β-catenin, and consequently mitigates osteosarcoma stemness. These discoveries position troxerutin as a promising candidate for targeted osteosarcoma therapy.
Collapse
Affiliation(s)
- Junkai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongbo Li
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qinglin Jin
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaoguang Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Gang Huang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Junqiang Yin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Changye Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinyu Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xin He
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Tiao Lin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
29
|
Zhang H, Wang Z, Qiao X, Wu J, Cheng C. Investigating potential drug targets for the treatment of glioblastoma: a Mendelian randomization study. BMC Cancer 2025; 25:654. [PMID: 40211130 PMCID: PMC11983800 DOI: 10.1186/s12885-025-13979-3] [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: 09/15/2024] [Accepted: 03/19/2025] [Indexed: 04/12/2025] Open
Abstract
Glioblastoma (GBM), one of the most aggressive brain tumors, has a 5-year survival rate of less than 5%. Current standard therapies, including surgery, radiotherapy, and temozolomide (TMZ) chemotherapy, are limited by drug resistance and the blood-brain barrier. Integrating expression quantitative trait loci (eQTL) and protein quantitative trait loci (pQTL) data has shown promise in uncovering disease mechanisms and therapeutic targets. This study combined eQTL and pQTL analyses to identify potential GBM-related genes and circulating plasma proteins for therapeutic exploration. Using transcriptomic data from The Cancer Genome Atlas (TCGA), we identified 2,528 differentially expressed genes, including GPX7 and CXCL10. eQTL-MR analysis identifies GBM-associated differentially expressed genes and constructs a protein-protein interaction (PPI) network.Integrating pQTL data from the deCODE database, pQTL-MR, and colocalization analyses validated the therapeutic potential of GPX7 and CXCL10.These findings provide new perspectives on GBM biology and suggest actionable targets for therapy. Despite limitations due to sample size and population-specific data, this study highlights GPX7 and CXCL10 as promising candidates for further investigation and lays the foundation for targeted GBM treatments.
Collapse
Affiliation(s)
- Hongwei Zhang
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui University of Science and Technology, Huainan, Anhui, 232001, China
| | - Zixuan Wang
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Xiaolong Qiao
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Jiaxing Wu
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Bengbu Medical University, Bengbu, Anhui, 233000, China
| | - Chuandong Cheng
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui University of Science and Technology, Huainan, Anhui, 232001, China.
| |
Collapse
|
30
|
Tang J, Amin MA, Campian JL. Glioblastoma Stem Cells at the Nexus of Tumor Heterogeneity, Immune Evasion, and Therapeutic Resistance. Cells 2025; 14:562. [PMID: 40277888 PMCID: PMC12025403 DOI: 10.3390/cells14080562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/26/2025] Open
Abstract
Glioblastoma (GBM) is an exceedingly aggressive primary brain tumor defined by rapid growth, extensive infiltration, and resistance to standard therapies. A central factor driving these malignancies is the subpopulation of glioblastoma stem cells (GSCs), which possess self-renewal capacity, multipotency, and the ability to regenerate tumor heterogeneity. GSCs contribute to key hallmarks of GBM pathobiology, including relentless progression, resistance to chemotherapy and radiotherapy, and inevitable recurrence. GSCs exhibit distinct molecular signatures, enhanced DNA repair, and metabolic adaptations that protect them against conventional treatments. Moreover, they reside within specialized niches-such as perivascular or hypoxic microenvironments-that sustain stemness, promote immunosuppression, and facilitate angiogenesis. Recent discoveries highlight signaling pathways like Notch, Wnt/β-catenin, Hedgehog, STAT3-PARN, and factors such as TFPI2 and HML-2 as critical regulators of GSC maintenance, plasticity, and immune evasion. These findings underscore the complexity of GSC biology and their pivotal role in driving GBM heterogeneity and therapeutic failure. Emerging therapeutic strategies aim to target GSCs through multiple avenues, including surface markers, immunotherapeutics (e.g., CAR T cells), metabolic vulnerabilities, and combination regimens. Advances in patient-derived organoids, single-cell omics, and 3D co-culture models enable more accurate representation of the tumor ecosystem and personalized therapeutic approaches. Ultimately, improved understanding of GSC-specific targets and the tumor microenvironment promises more effective interventions, paving the way toward better clinical outcomes for GBM patients.
Collapse
Affiliation(s)
- Justin Tang
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| | - Md Al Amin
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| | - Jian L. Campian
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| |
Collapse
|
31
|
Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025; 10:106. [PMID: 40180907 PMCID: PMC11968978 DOI: 10.1038/s41392-025-02142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 04/05/2025] Open
Abstract
The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination during development. Given its pivotal role in cellular communication, aberrant Wnt signaling has been extensively linked to the pathogenesis of various diseases. This review offers an in-depth analysis of the Wnt pathway, detailing its signal transduction mechanisms and principal components. Furthermore, the complex network of interactions between Wnt cascades and other key signaling pathways, such as Notch, Hedgehog, TGF-β, FGF, and NF-κB, is explored. Genetic mutations affecting the Wnt pathway play a pivotal role in disease progression, with particular emphasis on Wnt signaling's involvement in cancer stem cell biology and the tumor microenvironment. Additionally, this review underscores the diverse mechanisms through which Wnt signaling contributes to diseases such as cardiovascular conditions, neurodegenerative disorders, metabolic syndromes, autoimmune diseases, and cancer. Finally, a comprehensive overview of the therapeutic progress targeting Wnt signaling was given, and the latest progress in disease treatment targeting key components of the Wnt signaling pathway was summarized in detail, including Wnt ligands/receptors, β-catenin destruction complexes, and β-catenin/TCF transcription complexes. The development of small molecule inhibitors, monoclonal antibodies, and combination therapy strategies was emphasized, while the current potential therapeutic challenges were summarized. This aims to enhance the current understanding of this key pathway.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
32
|
Haddadin L, Sun X. Stem Cells in Cancer: From Mechanisms to Therapeutic Strategies. Cells 2025; 14:538. [PMID: 40214491 PMCID: PMC11988674 DOI: 10.3390/cells14070538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Stem cells have emerged as a pivotal area of research in the field of oncology, offering new insights into the mechanisms of cancer initiation, progression, and resistance to therapy. This review provides a comprehensive overview of the role of stem cells in cancer, focusing on cancer stem cells (CSCs), their characteristics, and their implications for cancer therapy. We discuss the origin and identification of CSCs, their role in tumorigenesis, metastasis, and drug resistance, and the potential therapeutic strategies targeting CSCs. Additionally, we explore the use of normal stem cells in cancer therapy, focusing on their role in tissue regeneration and their use as delivery vehicles for anticancer agents. Finally, we highlight the challenges and future directions in stem cell research in cancer.
Collapse
Affiliation(s)
| | - Xueqin Sun
- Cancer Genome and Epigenetics Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
33
|
Zheng W, Zhou C, Xue Z, Qiao L, Wang J, Lu F. Integrative analysis of a novel signature incorporating metabolism and stemness-related genes for risk stratification and assessing clinical outcomes and therapeutic responses in lung adenocarcinoma. BMC Cancer 2025; 25:591. [PMID: 40170009 PMCID: PMC11963273 DOI: 10.1186/s12885-025-13984-6] [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: 09/04/2024] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Metabolism and stemness-related genes (msRGs) are critical in the development and progression of lung adenocarcinoma (LUAD). Nevertheless, reliable prognostic risk signatures derived from msRGs have yet to be established. METHODS In this study, we downloaded and analyzed RNA-sequencing and clinical data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. We employed univariate and multivariate Cox regression analyses, along with least absolute shrinkage and selection operator (LASSO) regression analysis, to identify msRGs that are linked to the prognosis of LUAD and to develop the prognostic risk signature. The prognostic value was evaluated using Kaplan-Meier analysis and log-rank tests. We generated receiver operating characteristic (ROC) curves to evaluate the predictive capability of the prognostic signature. To estimate the relative proportions of infiltrating immune cells, we utilized the CIBERSORT algorithm and the MCPCOUNTER method. The prediction of the half-maximal inhibitory concentration (IC50) for commonly used chemotherapy drugs was conducted through ridge regression employing the "pRRophetic" R package. The validation of our analytical findings was performed through both in vivo and in vitro studies. RESULTS A novel five-gene prognostic risk signature consisting of S100P, GPX2, PRC1, ARNTL2, and RGS20 was developed based on the msRGs. A risk score derived from this gene signature was utilized to stratify LUAD patients into high- and low-risk groups, with the former exhibiting significantly poorer overall survival (OS). A nomogram was constructed incorporating the risk score and other clinical characteristics, showcasing strong capabilities in estimating the OS rates for LUAD patients. Furthermore, we observed notable differences in the infiltration of various immune cell subtypes, as well as in responses to immunotherapy and chemotherapy, between the low-risk and high-risk groups. Results from gene set enrichment analysis (GSEA) and in vitro studies indicated that the prognostic signature gene ARNTL2 influenced the prognosis of LUAD patients, primarily through the activation of the PI3K/AKT/mTOR signaling pathway. CONCLUSIONS Utilizing this gene signature for risk stratification could help with clinical treatment management and improve the prognosis of LUAD patients.
Collapse
Affiliation(s)
- Wanrong Zheng
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Chuchu Zhou
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Zixin Xue
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Ling Qiao
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jianjun Wang
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Feng Lu
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China.
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China.
| |
Collapse
|
34
|
Sailo BL, Chauhan S, Hegde M, Girisa S, Alqahtani MS, Abbas M, Goel A, Sethi G, Kunnumakkara AB. Therapeutic potential of tocotrienols as chemosensitizers in cancer therapy. Phytother Res 2025; 39:1694-1720. [PMID: 38353331 DOI: 10.1002/ptr.8131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/29/2023] [Accepted: 01/15/2024] [Indexed: 04/23/2025]
Abstract
Chemoresistance is the adaptation of cancer cells against therapeutic agents. When exhibited by cancer cells, chemoresistance helps them to avoid apoptosis, cause relapse, and metastasize, making it challenging for chemotherapeutic agents to treat cancer. Various strategies like dosage modification of drugs, nanoparticle-based delivery of chemotherapeutics, antibody-drug conjugates, and so on are being used to target and reverse chemoresistance, one among such is combination therapy. It uses the combination of two or more therapeutic agents to reverse multidrug resistance and improve the effects of chemotherapy. Phytochemicals are known to exhibit chemosensitizing properties and are found to be effective against various cancers. Tocotrienols (T3) and tocopherols (T) are natural bioactive analogs of vitamin E, which exhibit important medicinal value and potential curative properties apart from serving as an antioxidant and nutrient supplement. Notably, T3 exhibits a variety of pharmacological activities like anticancer, anti-inflammatory, antiproliferative, and so on. The chemosensitizing property of tocotrienol is exhibited by modulating several signaling pathways and molecular targets involved in cancer cell survival, proliferation, invasion, migration, and metastasis like NF-κB, STATs, Akt/mTOR, Bax/Bcl-2, Wnt/β-catenin, and many more. T3 sensitizes cancer cells to chemotherapeutic drugs including cisplatin, doxorubicin, and paclitaxel increasing drug concentration and cytotoxicity. Discussed herewith are the chemosensitizing properties of tocotrienols on various cancer cell types when combined with various drugs and biological molecules.
Collapse
Affiliation(s)
- Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Suravi Chauhan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Arul Goel
- University of California Santa Barbara, Santa Barbara, California, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
35
|
Huber K, Garg S, Schlautmann L, Wang R, He L, Huth R, Pouya A, Rohde C, Janssen M, Lüchtenborg C, Arnold C, Luque‐Navarro PM, Zaugg JB, Raffel S, Müller‐Tidow C, Jeremias I, López‐Cara LC, Brügger B, Pabst C. Phosphatidic acid phosphatase LPIN1 in phospholipid metabolism and stemness in hematopoiesis and AML. Hemasphere 2025; 9:e70118. [PMID: 40265168 PMCID: PMC12012646 DOI: 10.1002/hem3.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 12/14/2024] [Accepted: 02/12/2025] [Indexed: 04/24/2025] Open
Abstract
Targeting metabolism represents a promising approach to eradicate leukemic stem cells (LSCs) that are considered critical drivers of relapse in acute myeloid leukemia (AML). In this study, we demonstrate that the phosphatidic acid phosphatase LPIN1, which regulates the synthesis of diacylglycerol, the key substrate for triacylglycerol, and phospholipid production, is crucial for the function of healthy and leukemic hematopoietic stem and progenitor cells (HSPC and LSC). LPIN1 mRNA was highly expressed in the CD34+ compartment of primary human AML samples. LPIN1 suppression inhibited the proliferation of primary leukemic cells and normal HSPCs in vitro and in xenotransplantation assays. Lipidomics analyses revealed a reduction of phosphatidylcholine (PC) and phosphatidylethanolamine and an upregulation of sphingomyelin upon LPIN1 depletion. Distinct phospholipid composition was associated with genetic AML groups, and targeting PC production by choline kinase inhibitors showed strong anti-leukemic activity. In summary, our data establish a regulatory role of LPIN1 in HSPC and LSC function and provide novel insights into the role of glycerophospholipid homeostasis in stemness and differentiation.
Collapse
Affiliation(s)
- Karin Huber
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Department of Medicine II, Hematology and OncologyUniversity Hospital Schleswig HolsteinCampus KielGermany
| | - Swati Garg
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Department of Medical OncologyDana Farber Cancer InstituteBostonMassachusettsUSA
| | - Lena Schlautmann
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Rui Wang
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Lixiazi He
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Richard Huth
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Alireza Pouya
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Christian Rohde
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Maike Janssen
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | | | - Christian Arnold
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
- European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Pilar M. Luque‐Navarro
- Department of Pharmaceutical and Organic Chemistry, Faculty of PharmacyUniversity of GranadaGranadaSpain
| | - Judith B. Zaugg
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
- European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Simon Raffel
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Carsten Müller‐Tidow
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem CellsHelmholtz MunichOberschleißheimGermany
- Department of PediatricsDr. Von Hauner Children's Hospital, LMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK), Partner Site MunichMunichGermany
| | - Luisa C. López‐Cara
- Department of Pharmaceutical and Organic Chemistry, Faculty of PharmacyUniversity of GranadaGranadaSpain
| | - Britta Brügger
- Heidelberg University Biochemistry Center (BZH)HeidelbergGermany
| | - Caroline Pabst
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| |
Collapse
|
36
|
Li C, Zhang C, Li X. Clonal hematopoiesis of indeterminate potential: contribution to disease and promising interventions. Mol Cell Biochem 2025:10.1007/s11010-025-05261-8. [PMID: 40140229 DOI: 10.1007/s11010-025-05261-8] [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: 12/11/2024] [Accepted: 03/16/2025] [Indexed: 03/28/2025]
Abstract
In clonal hematopoiesis of indeterminate potential (CHIP), subpopulations of blood cells carrying somatic mutations expand as the individual ages, and this expansion may elevate risk of blood cancers as well as cardiovascular disease. Individuals at higher risk of CHIP and therefore of CHIP-associated disease can be identified through mutational profiling, and the apparently central role of inflammation in CHIP-associated disease has emerged as a potential therapeutic target. While CHIP is often associated with negative health outcomes, emerging evidence suggests that some CHIP-related mutations may also exert beneficial effects, indicating a more complex role in human health. This review examines current understanding of the epidemiology and clinical significance of CHIP and the role of inflammation in driving its association with disease risk. It explores the mechanisms linking CHIP to inflammation and risk of cardiovascular and other diseases, as well as the potential of personalizing therapies against those diseases for individuals with CHIP.
Collapse
Affiliation(s)
- Chongjie Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China
| | - Chunxiang Zhang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China.
| | - Xiuying Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
37
|
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
|
38
|
Almahdawi H, Akbas A, Rahebi J. Deep Learning Neural Network Based on PSO for Leukemia Cell Disease Diagnosis from Microscope Images. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01474-x. [PMID: 40113730 DOI: 10.1007/s10278-025-01474-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
Leukemia is a kind of cancer characterized by the proliferation of abnormal, immature White Blood Cells (WBCs) produced in the bone marrow, which subsequently circulate throughout the body. Prompt leukemia diagnosis is vital in determining the optimal treatment plan, as different types of leukemia require distinct treatments. Early detection is therefore instrumental in facilitating the use of the most effective therapies. The identification of leukemia cells from microscopic images is considered a challenging task due to the complexity of the image features. This paper presents a deep learning neural network approach that utilizes the Particle Swarm Optimization (PSO) method to diagnose leukemia cell disease from microscope images. Initially, deep learning is employed to extract features from the leukemia images, which are then optimized by the PSO method to select the most relevant features for machine learning. Three different machine learning algorithms, namely Decision Tree (DT), Support Vector Machine (SVM), and K-Nearest Neighbors (K-NN) methods, are utilized to analyze the selected features. The results of the experiments demonstrate PSO accuracies of 97.4%, 92.3%, and 85.9% for SVM, K-NN, and DT algorithms with GoogLeNet, respectively. The proposed method achieved accuracies of 100%, 94.9%, and 92.3% for SVM, K-NN, and DT methods respectively, with Ant Colony Optimization (ACO) feature extraction and ResNet-50 employed as revealed by the experimental results. These findings suggest that the proposed approach is a promising tool for accurate diagnosis of leukemia cell disease using microscopic images.
Collapse
Affiliation(s)
- Hamsa Almahdawi
- Computer Engineering Department, Cankiri Karatekin University, Cankiri, Turkey
| | - Ayhan Akbas
- Institute for Communication Systems, University of Surrey, Guildford, UK.
| | - Javad Rahebi
- Software Engineering Department, Istanbul Topkapi University, Istanbul, Turkey
| |
Collapse
|
39
|
Čikeš Botić M, Marijan S, Radan M, Novak I, Glumac M, Pilkington LI, Odak Z, Barker D, Reynisson J, Čikeš Čulić V. Thieno[2,3- b]pyridines as a Novel Strategy Against Cervical Cancer: Mechanistic Insights and Therapeutic Potential. Int J Mol Sci 2025; 26:2651. [PMID: 40141292 PMCID: PMC11942470 DOI: 10.3390/ijms26062651] [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: 01/31/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Cervical cancer is the fourth leading cause of cancer mortality in women worldwide, with limited therapeutic options for advanced or recurrent cases. In this study, the effects of a recent thieno[2,3-b]pyridine derivative, (E)-3-amino-5-(3-bromophenyl)acryloyl)-N-(3-chloro-2-methylphenyl)-6-methylthieno[2,3-b]pyridine-2-carboxamide (compound 1), on two cervical cancer cell lines, HeLa and SiHa, are investigated. Cytotoxicity was assessed by MTT assay, apoptosis rates were measured by flow cytometry, and metabolic profiling was performed by GC-MS. The study also examined the expression of eight glycosphingolipids (GSLs) in cancer stem cells (CSCs) and non-CSCs to assess glycophenotypic changes. Compound 1 showed significant cytotoxicity in both cell lines, with apoptosis identified as the primary mechanism of cell death. A significant reduction in the CSC population was observed, particularly in the SiHa cell line. Compound 1 treatment altered GSL expression and decreased GM2 levels in both CSCs and non-CSCs in the SiHa cell line and Gg3Cer levels in the HeLa cell line. Metabolic profiling identified 23 and 21 metabolites in the HeLa and SiHa cell lines, respectively, with significant differences in metabolite expression after treatment. These results underscore the potential of compound 1 as a promising therapeutic candidate for cervical cancer and warrant further investigation in preclinical and clinical settings.
Collapse
Affiliation(s)
- Monika Čikeš Botić
- Department of Gynecology and Obstetrics, University Hospital of Split, 21000 Split, Croatia;
| | - Sandra Marijan
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (S.M.); (V.Č.Č.)
| | - Mila Radan
- Department of Biochemistry, Faculty of Chemistry and Technology, University of Split, 21000 Split, Croatia;
| | - Ivana Novak
- Department of Immunology and Medical Genetics, School of Medicine, University of Split, 21000 Split, Croatia; (I.N.); (M.G.)
| | - Mateo Glumac
- Department of Immunology and Medical Genetics, School of Medicine, University of Split, 21000 Split, Croatia; (I.N.); (M.G.)
| | - Lisa I. Pilkington
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (L.I.P.); (D.B.)
- Te Pūnaha Matatini, Auckland 1042, New Zealand
| | - Zdravko Odak
- Department of Gynecology and Obstetrics, University Hospital of Split, 21000 Split, Croatia;
| | - David Barker
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (L.I.P.); (D.B.)
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6012, New Zealand
| | - Jóhannes Reynisson
- School of Allied Health Professions and Pharmacy, Keele University, Staffordshire ST5 5BG, UK;
| | - Vedrana Čikeš Čulić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (S.M.); (V.Č.Č.)
| |
Collapse
|
40
|
Xian W, Wang S, Xie J, Yamamoto Y, Khorrami M, Zhang Y, Montes RC, Desales C, Khorrami M, Mory Z, Hoffman A, Su A, Nguyen C, Davies PJA, Stephan C, Pan S, Wu W, Liu Y, Siegelman J, Waters RE, Ross WA, Song S, Metersky M, Beer DG, Crum CP, Stewart AJ, Vincent M, Russell R, Izard RA, Ho KY, Hung-Sen Lai J, Bachovchin WW, Ajani JA, McKeon FD. Evolution of Esophageal Adenocarcinoma From Precursor Lesion Stem Cells. Gastroenterology 2025:S0016-5085(25)00521-9. [PMID: 40090599 DOI: 10.1053/j.gastro.2025.02.032] [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: 11/07/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND AND AIMS Metastatic cancers arise from a decades-long succession of increasingly virulent precursor lesions, each of which represents prospective targets for therapeutic intervention. This evolutionary process has been particularly vivid in esophageal adenocarcinoma (EAC), as this cancer and associated precursor lesions, including Barrett's esophagus (BE), low-grade dysplasia (LGD), and high-grade dysplasia (HGD), coexist in an accessible, 2-dimensional pattern in esophageal mucosa. Given the durability of these precursor lesions, it is likely that they, like EAC, rely on stem cells for their regenerative growth. To assess the role of stem cells in the evolution of EAC, we apply technology that selectively clones stem cells from the gastrointestinal tract to patient-matched endoscopic biopsies from each of the precursor lesions implicated in EAC. METHODS Histologically validated, endoscopic biopsy series including EAC, HGD, LGD, BE, and normal esophageal mucosa were obtained from patients presenting with EAC. Rare (1:1000) cells from each of these lesions proved clonogenic and were assessed by in vitro differentiation, tumorigenicity in mice, and by molecular genetics. RESULTS Each of the lesions in the evolution of EAC possesses a discrete set of clonogenic cells marked by immaturity, enormous proliferative potential, and lesion-specific differentiation fate. DNA sequencing of these clones reveals intralesional heterogeneity and clonal resolution of the mutation progression within a given patient from BE, LGD, HGD, and EAC. High-throughput chemical screens against BE stem cells reveal drug combinations that are similarly effective against stem cells of LGD, HGD, and EAC. CONCLUSIONS All lesions in the evolution of EAC possess discrete populations of stem cells that are potential therapeutic targets.
Collapse
Affiliation(s)
- Wa Xian
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Shan Wang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Jingzhong Xie
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Yusuke Yamamoto
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Melina Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Yanting Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | - Caycel Desales
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Melika Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Zaal Mory
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Ashley Hoffman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Amber Su
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Crystal Nguyen
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | | | - Shuang Pan
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Wengen Wu
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Yuxin Liu
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Jeremy Siegelman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Rebecca E Waters
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William A Ross
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark Metersky
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - David G Beer
- Departments of Thoracic Surgery and Radiation Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Christopher P Crum
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Alexander J Stewart
- School of Mathematics and Statistics, University of St. Andrews, North Haugh, UK
| | | | | | | | - Khek Yu Ho
- Department of Medicine, National University of Singapore, Singapore
| | - Jack Hung-Sen Lai
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts; Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts
| | - William W Bachovchin
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts; Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frank D McKeon
- Department of Biology and Biochemistry, University of Houston, Houston, Texas.
| |
Collapse
|
41
|
Aoki Y, Wang L, Tsuda M, Saito Y, Kubota T, Oda Y, Hirano S, Gong JP, Tanaka S. Hydrogel PCDME creates pancreatic cancer stem cells in OXPHOS metabolic state with TXNIP elevation. Biochem Biophys Res Commun 2025; 751:151416. [PMID: 39914146 DOI: 10.1016/j.bbrc.2025.151416] [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: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
Pancreatic cancer is known as one of the poor prognostic cancers, and the most of patients are unable to undergo radical resection due to local progression or distant metastasis at initial diagnosis. In spite of the advancements in surgery and chemotherapy, there are many cases of recurrence after surgery or chemoradiotherapy mainly due to the presence of cancer stem cells (CSCs). CSCs are potential therapeutic target, but current issue is that an identification of CSCs is difficult since they are only present in a small number of tumor cells. Here we demonstrate that hydrogel PCDME can rapidly induce pancreatic cancer cell spheroids with elevated levels of stem cell markers including Sox2, Oct3/4, and Nanog, and the growth rate was reduced. CSCs showed activation of YAP/TAZ signaling, and microarray analysis showed markedly elevated expression of thioredoxin-interacting protein (TXNIP). Primary pancreatic cancer cells also increased TXNIP in addition to stemness markers on gel. In metabolic analysis, CSCs showed a shift of energy production from glycolysis to oxidative phosphorylation (OXPHOS). Furthermore, knockdown of TXNIP on PCDME gel using shRNAs decreased growth speed and in vivo tumorigenicity, suggesting that TXNIP may be involved in CSCs induction.
Collapse
Affiliation(s)
- Yuma Aoki
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Yusuke Saito
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Takenori Kubota
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Jian Ping Gong
- World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan; Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan; World Premier International Research Center Initiative, Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan; Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan.
| |
Collapse
|
42
|
Rouault CD, Bansard L, Martínez-Balsalobre E, Bonnet C, Wicinski J, Lin S, Colombeau L, Debieu S, Pinna G, Vandamme M, Machu M, Rosnet O, Chevrier V, Popovici C, Sobol H, Castellano R, Pasquier E, Guasch G, Rodriguez R, Pannequin J, Pascussi JM, Lachaud C, Charafe-Jauffret E, Ginestier C. Inhibition of the STAT3/Fanconi anemia axis is synthetic lethal with PARP inhibition in breast cancer. Nat Commun 2025; 16:2159. [PMID: 40038300 PMCID: PMC11880418 DOI: 10.1038/s41467-025-57476-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025] Open
Abstract
The targeting of cancer stem cells (CSCs) has proven to be an effective approach for limiting tumor progression, thus necessitating the identification of new drugs with anti-CSC activity. Through a high-throughput drug repositioning screen, we identify the antibiotic Nifuroxazide (NIF) as a potent anti-CSC compound. Utilizing a click chemistry strategy, we demonstrate that NIF is a prodrug that is specifically bioactivated in breast CSCs. Mechanistically, NIF-induced CSC death is a result of a synergistic action that combines the generation of DNA interstrand crosslinks with the inhibition of the Fanconi anemia (FA) pathway activity. NIF treatment mimics FA-deficiency through the inhibition of STAT3, which we identify as a non-canonical transcription factor of FA-related genes. NIF induces a chemical HRDness (Homologous Recombination Deficiency) in CSCs that (re)sensitizes breast cancers with innate or acquired resistance to PARP inhibitor (PARPi) in patient-derived xenograft models. Our results suggest that NIF may be useful in combination with PARPi for the treatment of breast tumors, regardless of their HRD status.
Collapse
Affiliation(s)
- Celia D Rouault
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France
| | - Lucile Bansard
- IGF, University Montpellier, CNRS INSERM, Montpellier, France
| | - Elena Martínez-Balsalobre
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, DNA Interstrand Crosslink Lesions and Blood Disorder Team, Marseille, France
| | - Caroline Bonnet
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France
| | - Julien Wicinski
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France
| | - Shuheng Lin
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France
| | - Ludovic Colombeau
- Institut Curie, CNRS, INSERM, Biomedicine Laboratory PSL Research University, Paris, France
| | - Sylvain Debieu
- Institut Curie, CNRS, INSERM, Biomedicine Laboratory PSL Research University, Paris, France
| | - Guillaume Pinna
- Plateforme ARN Interférence (PARI), Université Paris Cité, Inserm, CEA Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Marie Vandamme
- Plateforme ARN Interférence (PARI), Université Paris Cité, Inserm, CEA Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Margot Machu
- IGF, University Montpellier, CNRS INSERM, Montpellier, France
| | - Olivier Rosnet
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France
| | - Véronique Chevrier
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France
| | - Cornel Popovici
- Aix-Marseille University, Cancer Genetic Laboratory, Cancer Biology Department Institut Paoli-Calmettes, Marseille, France
| | - Hagay Sobol
- Aix-Marseille University, Cancer Genetic Laboratory, Cancer Biology Department Institut Paoli-Calmettes, Marseille, France
| | - Rémy Castellano
- CRCM, Aix-Marseille University, INSERM, CNRS, Institut Paoli-Calmettes, TrGET Plateform, Marseille, France
| | - Eddy Pasquier
- CRCM, INSERM, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Reverse Molecular Pharmacology in Pediatric Oncology, Marseille, France
| | - Geraldine Guasch
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France
| | - Raphaël Rodriguez
- Institut Curie, CNRS, INSERM, Biomedicine Laboratory PSL Research University, Paris, France
| | - Julie Pannequin
- IGF, University Montpellier, CNRS INSERM, Montpellier, France
| | | | - Christophe Lachaud
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, DNA Interstrand Crosslink Lesions and Blood Disorder Team, Marseille, France
| | - Emmanuelle Charafe-Jauffret
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France.
| | - Christophe Ginestier
- CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, Epithelial Stem Cells and Cancer Lab, Equipe Labellisée LIGUE Contre Le Cancer, Marseille, France.
| |
Collapse
|
43
|
Giron-Michel J, Padelli M, Oberlin E, Guenou H, Duclos-Vallée JC. State-of-the-Art Liver Cancer Organoids: Modeling Cancer Stem Cell Heterogeneity for Personalized Treatment. BioDrugs 2025; 39:237-260. [PMID: 39826071 PMCID: PMC11906529 DOI: 10.1007/s40259-024-00702-0] [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] [Accepted: 12/21/2024] [Indexed: 01/20/2025]
Abstract
Liver cancer poses a global health challenge with limited therapeutic options. Notably, the limited success of current therapies in patients with primary liver cancers (PLCs) may be attributed to the high heterogeneity of both hepatocellular carcinoma (HCCs) and intrahepatic cholangiocarcinoma (iCCAs). This heterogeneity evolves over time as tumor-initiating stem cells, or cancer stem cells (CSCs), undergo (epi)genetic alterations or encounter microenvironmental changes within the tumor microenvironment. These modifications enable CSCs to exhibit plasticity, differentiating into various resistant tumor cell types. Addressing this challenge requires urgent efforts to develop personalized treatments guided by biomarkers, with a specific focus on targeting CSCs. The lack of effective precision treatments for PLCs is partly due to the scarcity of ex vivo preclinical models that accurately capture the complexity of CSC-related tumors and can predict therapeutic responses. Fortunately, recent advancements in the establishment of patient-derived liver cancer cell lines and organoids have opened new avenues for precision medicine research. Notably, patient-derived organoid (PDO) cultures have demonstrated self-assembly and self-renewal capabilities, retaining essential characteristics of their respective in vivo tissues, including both inter- and intratumoral heterogeneities. The emergence of PDOs derived from PLCs serves as patient avatars, enabling preclinical investigations for patient stratification, screening of anticancer drugs, efficacy testing, and thereby advancing the field of precision medicine. This review offers a comprehensive summary of the advancements in constructing PLC-derived PDO models. Emphasis is placed on the role of CSCs, which not only contribute significantly to the establishment of PDO cultures but also faithfully capture tumor heterogeneity and the ensuing development of therapy resistance. The exploration of PDOs' benefits in personalized medicine research is undertaken, including a discussion of their limitations, particularly in terms of culture conditions, reproducibility, and scalability.
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France.
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France.
| | - Maël Padelli
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- Department of Biochemistry and Oncogenetics, Paul Brousse Hospital, AP-HP, Villejuif, France
| | - Estelle Oberlin
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Hind Guenou
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Jean-Charles Duclos-Vallée
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- INSERM UMR-S 1193, Paul Brousse Hospital, Villejuif, France
- Hepato-Biliary Department, Paul Brousse Hospital, APHP, Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hepatinov, Villejuif, France
| |
Collapse
|
44
|
Zhu W, Wang J, Lei K, Yan X, Xu J, Liu S, Li C. Leading edge biosensing applications based on AIE technology. Biosens Bioelectron 2025; 271:116953. [PMID: 39622156 DOI: 10.1016/j.bios.2024.116953] [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: 06/06/2024] [Revised: 11/04/2024] [Accepted: 11/16/2024] [Indexed: 01/06/2025]
Abstract
Luminescent materials provide a unique method for biological imaging. Luminescent probes can label molecules of interest and present luminescent signals. Bioluminescence bioimaging has shown great efficacy in environmental, live cell and animal studies. Light-emitting materials play a very wide role in the field of light-emitting devices and biosensing. Luminescent materials are usually used as solid films or aggregate states. However, it is difficult to monitor the selectivity and sensitivity of various ions and small molecules in living cells with ordinary luminescent materials due to the changes in various aspects of analytes. Organic luminescent materials exhibit aggregation-induced quenching (ACQ) on molecular aggregation, and the ACQ effect is very common, which greatly limits the application of luminescent materials in chemical sensing, especially in biological imaging. Academician Tang Benzhong proposed "aggregation-induced emission (AIE)" as a powerful method to solve the ACQ problem for the first time. In this paper, the working principle of AIE is reviewed, and the research on the core working mechanism of AIE technology is not only of great fundamental significance, but also can pave the way for practical innovation of AIE technology applications. In this review, we outline the current basic understanding of the working mechanism of AIE, collate the cutting-edge biosensing applications based on AIE technology, including applications based on AIE in substance detection, biological detection, and disease detection. At last, we discuss the future development of AIE research.
Collapse
Affiliation(s)
- Weitao Zhu
- Clinical Medicine (Eight-Year Program), West China School of Medicine, Sichuan University, Chengdu, 610044, China
| | - Jiaao Wang
- Clinical Medicine (Eight-Year Program), West China School of Medicine, Sichuan University, Chengdu, 610044, China
| | - Kaixin Lei
- Clinical Medicine (Eight-Year Program), West China School of Medicine, Sichuan University, Chengdu, 610044, China
| | - Xu Yan
- State Key Laboratory of Integrated Optoelectronics, Key Laboratory of Advanced Gas Sensors, Jilin Province, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, China
| | - Jiancheng Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun, 130021, China
| | - Shan Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Chenzhong Li
- Biomedical Engineering, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Juxintang (Chengdu) Biotechnology Co., Ltd., Chengdu, 641400, China
| |
Collapse
|
45
|
Chen L, Fang R, Cai Z, Huang B, Zhang J, Li Y, Chen Y, Xu Z, Lei W, Zhang M. CD271 high cancer stem cells regulate macrophage polarization in head and neck squamous cell carcinoma. Oral Oncol 2025; 162:107181. [PMID: 39854870 DOI: 10.1016/j.oraloncology.2025.107181] [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: 11/04/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
PURPOSE Cancer stem cells (CSCs) are considered key drivers of progression in head and neck squamous cell carcinoma (HNSCC). Our single-cell RNA sequencing (scRNA-seq) analysis revealed predominant expression of CD271 in CSCs, however, its role as a CSC marker in HNSCC requires further elucidation. We investigated the stemness characteristics of CD271high HNSCC cells and their interactions with the tumor immune microenvironment. METHODS scRNA-seq data from hypopharyngeal squamous cell carcinoma (HPSCC) tissues were analyzed to identify expression profile of CSCs. Overall survival was compared between CD271high and CD271low patients based on immunostaining of HPSCC samples. The stemness of CD271high HNSCC cells was evaluated via an in vivo limiting dilution assay. In a C57BL/6 mice model, the percentage of immune cells and macrophage subtypes were analyzed by flow cytometry. The role of CD271 in macrophage polarization was further examined by in vitro coculture of CD271high cells with CD14+ monocytes. Gene expressions were analyzed by qPCR. RESULTS CD271 is predominantly expressed in CSCs identified by scRNA-seq analysis. CD271 enhances HNSCC cell proliferation and is negatively correlated with patient prognosis in HPSCC. CD271 knockdown suppressed HNSCC tumor growth and regulated macrophage polarization within the TME. CD271high cells exhibited stemness features and enhanced tumor growth in vivo. CONCLUSIONS CD271high HNSCC cells exhibit CSC characteristics and regulate macrophage polarization. Targeting CD271 may improve the immunosuppressive TME to inhibit tumor growth. Combining CD271-targeting agents with other therapies presents a promising strategy that may enhance therapeutic efficacy and prognosis in HNSCC.
Collapse
Affiliation(s)
- Lifan Chen
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Ruihua Fang
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Zhimou Cai
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Bixue Huang
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Jinhong Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Yun Li
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Yi Chen
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Zhenglin Xu
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China
| | - Wenbin Lei
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China.
| | - Minjuan Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, PR China.
| |
Collapse
|
46
|
Yang L, Yi Y, Mei Z, Huang D, Tang S, Hu L, Liu L. Circular RNAs in cancer stem cells: Insights into their roles and mechanisms (Review). Int J Mol Med 2025; 55:50. [PMID: 39930823 PMCID: PMC11781527 DOI: 10.3892/ijmm.2025.5491] [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: 09/27/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025] Open
Abstract
Cancer stem cells (CSCs) represent a small, yet pivotal subpopulation of tumor cells that play significant roles in tumor initiation, progression and therapeutic resistance. Circular RNAs (circRNAs) are a distinct class of RNAs characterized by their closed‑loop structures, lacking 5' to 3'ends. There is growing evidence that circRNAs are integral to the development and regulation of CSCs. Aberrant expression of circRNAs in CSCs can contribute to oncogenic properties and drug resistance. Specifically, oncogenic circRNAs modulate CSC behavior via key signaling pathways, thereby promoting CSC self‑renewal and maintenance, as well as tumor progression. This review summarizes the latest research on the functional roles and regulatory mechanisms of circRNAs in CSC behavior and discusses potential applications and challenges of targeting circRNAs in CSCs. Understanding the intricate interactions between circRNAs and CSCs may lead to novel therapeutic strategies that effectively combat treatment resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Lunyu Yang
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Yuling Yi
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Zhu Mei
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Dongmei Huang
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Sitian Tang
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Liyi Hu
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| | - Ling Liu
- Department of Medical Laboratory, Chongqing Liangjiang New Area People's Hospital, Chongqing 401121, P.R. China
| |
Collapse
|
47
|
Gao R, Chen K, Wang Y, Guo R, Zhang X, Wu P, Wang W, Huang Q, Xie X, Yang S, Lv Y, Ren Q, Liu F, Chen S, Ma F, Cheng T, Cheng H. FHL2 deficiency aggravates Candida albicans infection through decreased myelopoiesis. SCIENCE CHINA. LIFE SCIENCES 2025; 68:722-733. [PMID: 39815033 DOI: 10.1007/s11427-024-2645-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/06/2024] [Indexed: 01/18/2025]
Abstract
Hematopoiesis is a finely tuned process that generates all blood cell types through self-renewal and differentiation, which is crucial for maintaining homeostasis. Acute infections can prompt a hematopoietic response known as emergency myelopoiesis. In this study, using a Candida albicans (C. albicans) infection model, we demonstrated for the first time that disruption of Fhl2 led to increased fungal burden, heightened inflammatory response and reduced survival rates. Impaired myeloid hematopoiesis and immune cell production were evident, as proved by the decreased numbers of hematopoietic stem and progenitor cells (HSPCs) and granulocytes in the bone marrow of Fhl2-deficient mice. In conclusion, FHL2 regulated emergency myelopoiesis in response to C. albicans, affecting the host's defense against pathogens.
Collapse
Affiliation(s)
- Rongmei Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Kanchao Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Yimin Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Rongxia Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Xiaoyu Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Weili Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Qingxiang Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Xuemei Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Shangda Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Yanling Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Fei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Song Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Fengxia Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China.
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- PUMC Department of Stem Cell and Regenerative Medicine, CAMS Center for Stem Cell Medicine, Tianjin, 300020, China.
| |
Collapse
|
48
|
Chin-Yee B, Laplane L, Sujobert P. Epistemic limitations of measurable residual disease in haematological malignancies. Lancet Haematol 2025; 12:e224-e229. [PMID: 40044257 DOI: 10.1016/s2352-3026(25)00002-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/30/2024] [Accepted: 01/08/2025] [Indexed: 05/13/2025]
Abstract
The growing use of measurable residual disease (MRD) assays across haematology-oncology creates an urgent need for clinicians and researchers to reflect on the biological and clinical rationale of this class of biomarkers. In this Viewpoint, we critically examine two premises behind MRD's use in haematology-oncology, focusing on its biological plausibility as a predictive biomarker and surrogate endpoint, and the evidence needed for it to influence decision making in haematological cancers. Examining these premises leads us to advocate for the establishment of more robust biological and clinical evidence to ensure the clinically useful and safe application of MRD. Although achieving the eradication of cancer cells in the form of undetectable MRD seems an attractive goal in haematology-oncology, we highlight the epistemic limitations of this biomarker and need for more clinical evidence to guide its effective use.
Collapse
Affiliation(s)
- Benjamin Chin-Yee
- Department of Pathology and Laboratory Medicine, and Division of Hematology, Department of Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON, Canada; Department of History and Philosophy of Science, University of Cambridge, Cambridge, UK
| | - Lucie Laplane
- CNRS, Institut d'Histoire et Philosophie des Sciences et des Techniques, University Paris 1 Panthéon-Sorbonne, Paris, France; Institut Gustave Roussy, Hematopoietic stem cells and the development of myeloid malignancies, Villejuif, France
| | - Pierre Sujobert
- Centre International de Recherche en Infectiologie (INSERM U1111, CNRS UMR 5308, École Normale supérieure de Lyon), Lymphoma ImmunoBiology team, Faculté de Médecine Lyon sud, Université Claude Bernard Lyon 1, Lyon, France; Service d'hématologie biologique, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre Bénite, France.
| |
Collapse
|
49
|
Joshi H, Yavuz M, Taylan O, Alkabaa A. Dynamic analysis of fractal-fractional cancer model under chemotherapy drug with generalized Mittag-Leffler kernel. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 260:108565. [PMID: 39732085 DOI: 10.1016/j.cmpb.2024.108565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/04/2024] [Accepted: 12/14/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND AND OBJECTIVE Cancer's complex and multifaceted nature makes it challenging to identify unique molecular and pathophysiological signatures, thereby hindering the development of effective therapies. This paper presents a novel fractal-fractional cancer model to study the complex interplay among stem cells, effectors cells, and tumor cells in the presence and absence of chemotherapy. The cancer model with effective treatment through chemotherapy drugs is considered and discussed in detail. METHODS The numerical method for the fractal-fractional cancer model with a generalized Mittag-Leffler kernel is presented. The Routh-Hurwitz stability criteria are applied to confirm the local asymptotically stability of an endemic equilibrium point of the cancer model without treatment and with effective treatment under some conditions. The existence and uniqueness criteria of the fractal-fractional cancer model are derived. Furthermore, the stability analysis of the fractal-fractional cancer model is performed. RESULTS The temporal concentration pattern of stem cells, effectors cells, tumor cells, and chemotherapy drugs are procured. The usage of chemotherapy drugs kills the tumor cells or decreases their density over time and as a consequence takes a longer time to reach to equilibrium point. The decay rate of stem cells and tumor cells plays a crucial role in cancer dynamics. The notable role of fractal dimensions along with fractional order is observed in capturing the cancer cell concentration. CONCLUSION A dynamic analysis of the fractal-fractional cancer model is demonstrated to examine the impact of chemotherapy drugs with a generalized Mittag-Leffler kernel. The model possesses three equilibrium points among them two correspond to the cancer model without treatment namely the tumor-free equilibrium point and endemic equilibrium point. One additional endemic equilibrium point exists in the case of effective treatment through chemotherapy drugs. The Routh-Hurwitz stability criteria are applied to confirm the local asymptotically stability of an endemic equilibrium point of the cancer model with and without treatment under some conditions. The chemotherapy drug plays a crucial role in controlling the growth of tumor cells. The fractal-fractional operator provided robustness to study cancer dynamics by the inclusion of memory and heterogeneity.
Collapse
Affiliation(s)
- Hardik Joshi
- Department of Mathematics, LJ Institute of Engineering and Technology, LJ University, Ahmedabad, 382210, Gujarat, India.
| | - Mehmet Yavuz
- Department of Mathematics and Computer Sciences, Faculty of Science, Necmettin Erbakan University, Konya 42090, Turkiye; Department of Applied Mathematics and Informatics, Kyrgyz-Turkish Manas University, Bishkek 720038, Kyrgyzstan.
| | - Osman Taylan
- Department of Management Engineering, Faculty of Management, Istanbul Technical University, Istanbul, Macka, Turkiye.
| | - Abdulaziz Alkabaa
- Department of Industrial Engineering, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
50
|
ZHOU DAIEN, YUAN HAOYANG, HU YIWEI, WANG CHUXU, GE SA, SHAO KOUFENG, WANG HONGYING, TIAN XIAOFENG, HU HAIBO. Loss of TNFRSF21 induces cisplatin sensitivity in lung adenocarcinoma. Oncol Res 2025; 33:653-663. [PMID: 40109864 PMCID: PMC11915077 DOI: 10.32604/or.2024.050182] [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: 01/30/2024] [Accepted: 06/07/2024] [Indexed: 03/22/2025] Open
Abstract
Background Despite the identification of numerous therapeutic targets in lung cancer, achieving significant efficacy has been challenging. TNFRSF21 plays an important role in various cancers. We investigated the function of TNFRSF21 in lung adenocarcinoma (LUAD). Methods The prognostic value of TNFRSF21 expression in lung cancer was evaluated by the GEPIA and Kaplan-Meier Plotter databases. Lung cancer cell viability was assessed by the CCK8 assay. TNFRSF21 expression patterns in lung cancer tissues and cells were examined using RT-PCR assay. Tumor sphere growth was evaluated through tumor sphere formation assays. MtROS contents in lung cancer cells were observed through MitoSOX fluorescent assays. Result TNFRSF21 was up-regulated in LUAD patients. TNFRSF21 induction was particularly notable in LUAD, especially in cancerous cells (A549, H1299, H460, and SPC-A1), compared to BEAS-2B cells. Additionally, TNFRSF21 was increased in cisplatin (DDP)-resistant LUAD cells. Loss of TNFRSF21 significantly inhibited LUAD cell growth. It was observed that forced expression of TNFRSF21 contributed to tumor cell proliferation and DDP resistance. The production of ROS was found to participate in the inhibitory effects on lung cancer stem cells (CSCs), with decreased TNFRSF21 restraining ROS contents. Collectively, these findings reveal that the downregulation of TNFRSF21 promotes ROS contents to restrain the lung CSC-like characteristics via modulation of CD44 and CD133. Conclusions In conclusion, TNFRSF21 may act as a novel target for lung cancer chemotherapy, particularly for eradicating lung CSCs.
Collapse
Affiliation(s)
- DAIEN ZHOU
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - HAOYANG YUAN
- Medical Faculty, Kunming University of Science and Technology, Kunming, 650000, China
| | - YIWEI HU
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - CHUXU WANG
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - SA GE
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - KOUFENG SHAO
- Department of Oncology, Huai’an Chuzhou Hospital of Traditional Chinese Medicine, Zhongda Hospital Group Hospital Affiliated to Southeast University, Huai’an, 223001, China
| | - HONGYING WANG
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - XIAOFENG TIAN
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| | - HAIBO HU
- Department of Thoracic Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, 223001, China
| |
Collapse
|