1
|
Zhou S, Jiang D, Liu Y, Wang Q, Hu M, Dai K, Chen L, Zhang T, Cai C, Wang J. The role of Sine Oculis Homeobox Homolog 2 in colon Cancer: Insights into prognosis, immune regulation, and therapeutic implications. Biochem Biophys Res Commun 2025; 772:152038. [PMID: 40414014 DOI: 10.1016/j.bbrc.2025.152038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/19/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Colon cancer (CC) remains a significant global health burden, and the search for novel prognostic biomarkers and therapeutic targets is crucial. This study comprehensively analyzed the role of SIX2 (Sine Oculis Homeobox Homolog 2) in CC. Utilizing data from TCGA, GTEx, and CCLE databases, differential expression of SIX2 was observed in multiple cancers, with significant upregulation in many tumors compared to normal tissues. In CC, SIX2's differential expression was notable. Cox regression analysis revealed its prognostic significance, with overexpression associated with poor survival outcomes. SIX2 was strongly associated with gene alterations and correlated with key signaling pathways like WNT and TGF-β. In the tumor microenvironment, SIX2 was related to immune cell infiltration and immune-related molecules. Notably, in CC, it was associated with immunosuppressive cells and checkpoint molecules. Additionally, ABT737 was found to sensitize tumor immunotherapy in the context of SIX2. Animal experiments demonstrated that ABT737 effectively restricted the growth of CC in mice, and its combination with antiPD-1 immunotherapy was more effective. It could reduce the infiltration of CD163+ tumor-associated macrophages but without significantly increasing the infiltration of CD8+ T cells. Our findings suggest that SIX2 is a potential key player in CC, offering insights into future research and the development of targeted therapies.
Collapse
Affiliation(s)
- Shicheng Zhou
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Dan Jiang
- Department of Surgery, Wucheng District People's Hospital, Jinhua, Zhejiang, 321000, China
| | - Yu Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qin Wang
- Department of Endocrinology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Manyi Hu
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Kangfu Dai
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Lin Chen
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China
| | - Tianming Zhang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Cheng Cai
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China.
| | - Jianping Wang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China.
| |
Collapse
|
2
|
Rahimi M, Kariminezhad Z, Rondon EP, Fahmi H, Fernandes JC, Benderdour M. Chitosan nanovectors for siRNA delivery: New horizons for nonviral gene therapy. Carbohydr Polym 2025; 360:123581. [PMID: 40399008 DOI: 10.1016/j.carbpol.2025.123581] [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: 01/04/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 05/23/2025]
Abstract
The growing interest in RNA-based therapeutics has positioned small interfering RNA (siRNA) as a promising tool for gene silencing with high specificity and efficacy. However, the successful clinical application of siRNA therapies requires efficient delivery systems to overcome extracellular and intracellular barriers. Chitosan, a naturally derived polysaccharide, has gained significant attention as a non-viral vector due to its biodegradability, biocompatibility, mucoadhesive properties, and capacity to enhance cellular uptake. These attributes make chitosan an attractive alternative to lipid-based nanoparticles, which currently dominate siRNA delivery platforms. Recent advancements in chitosan-based nanoformulations, including chemical modifications and functionalization strategies, have improved siRNA stability, targeting efficiency, and transfection potential, addressing key limitations such as low bioavailability and immunogenicity. Despite these advances, challenges remain in achieving optimal release kinetics, scalability, and consistent therapeutic efficacy. Future research efforts will focus on engineering chitosan derivatives with enhanced physicochemical properties, integrating multifunctional nanocarriers, and refining formulation strategies to bridge the gap between preclinical research and clinical translation. The continued development of chitosan-based siRNA therapeutics holds significant potential for advancing precision medicine and expanding treatment options for a variety of diseases, including cancer, metabolic disorders, and inflammatory conditions.
Collapse
Affiliation(s)
- Mahdi Rahimi
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada
| | - Zahra Kariminezhad
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada; Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Elsa-Patricia Rondon
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada; Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Julio C Fernandes
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada; Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Mohamed Benderdour
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada.
| |
Collapse
|
3
|
Gupta MK, Gouda G, Moazzam-Jazi M, Vadde R, Nagaraju GP, El-Rayes BF. CRISPR/Cas9-directed epigenetic editing in colorectal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189338. [PMID: 40315964 DOI: 10.1016/j.bbcan.2025.189338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/21/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related illness and death worldwide, arising from a complex interplay of genetic predisposition, environmental influences, and epigenetic dysregulation. Among these factors, epigenetic modifications-reversible and heritable changes in gene expression-serve as crucial regulators of CRC progression. Understanding these modifications is essential for identifying potential biomarkers for early diagnosis and developing targeted therapeutic strategies. Epigenetic drugs (epidrugs) such as DNA methyltransferase inhibitors (e.g., decitabine) and bromodomain inhibitors (e.g., JQ1) have shown promise in modulating aberrant epigenetic changes in CRC. However, challenges such as drug specificity, delivery, and safety concerns limit their clinical application. Advances in CRISPR-Cas9-based epigenetic editing offer a more precise approach to modifying specific epigenetic markers, presenting a potential breakthrough in CRC treatment. Despite its promise, CRISPR-based epigenome editing may result in unintended genetic modifications, necessitating stringent regulations and safety assessments. Beyond pharmacological interventions, lifestyle factors-including diet and gut microbiome composition-play a significant role in shaping the epigenetic landscape of CRC. Nutritional and microbiome-based interventions have shown potential in preventing CRC development by maintaining intestinal homeostasis and reducing tumor-promoting epigenetic changes. This review provides a comprehensive overview of epigenetic alterations in CRC, exploring their implications for diagnosis, prevention, and treatment. By integrating multi-omics approaches, single-cell technologies, and model organism studies, future research can enhance the specificity and efficacy of epigenetic-based therapies. Shortly, a combination of advanced gene-editing technologies, targeted epidrugs, and lifestyle interventions may pave the way for more effective and personalized CRC treatment strategies.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover 30625, Germany
| | - Gayatri Gouda
- ICAR-National Rice Research Institute, Cuttack 753 006, Odisha, India
| | - Maryam Moazzam-Jazi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramakrishna Vadde
- Department of Biotechnology and Bioinformatics, Yogi Vemana University, Kadapa 516005, Andhra Pradesh, India
| | - Ganji Purnachandra Nagaraju
- Division of Hematology & Oncology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Bassel F El-Rayes
- Division of Hematology & Oncology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
4
|
Fang X, Zhao J, Wu S, Liao P, Guan G. The intestinal toxicity mechanisms of triclosan and triclocarban and their possible clinical nutritional intervention mechanisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126396. [PMID: 40345375 DOI: 10.1016/j.envpol.2025.126396] [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: 03/13/2025] [Revised: 04/20/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Triclosan (TCS) and triclocarban (TCC) are widely used as antimicrobial agents in personal care products. Their widespread use has become a potential environmental contaminant. This review reviews the mechanisms of intestinal toxicity of TCS and TCC and their potential nutritional intervention strategies. TCS and TCC can be metabolized to glucuronic acid conjugates in the host and subsequently uncoupled by microorganisms in the intestine to regenerate free forms of TCS and TCC. TCS and TCC are unique metabolic pathways that lead to accumulation in the gut, altering the structure of intestinal flora, increasing the relative abundance of pathogenic bacteria, while reducing the abundance of beneficial bacteria, thereby disrupting the balance of intestinal flora. In addition, they can interfere with the self-renewal and differentiation of ISCs, thereby weakening intestinal barrier function. TCS and TCC can also activate the TLR4-NFκB signaling pathway, inducing and exacerbating inflammatory responses. These mechanisms together lead to intestinal toxicity and have a significant negative impact on intestinal health. In order to cope with the intestinal toxicity caused by these mechanisms of action, this paper believes that prebiotics, probiotics, vitamins, minerals and herbal extracts can be used as potential nutritional interventions to reduce the intestinal toxicity of TCS and TCC by regulating intestinal microbiota, enhancing intestinal barrier function and inhibiting inflammatory response. Although preliminary studies have shown the potential benefits of these interventions, their specific efficacy and safety still need further study.
Collapse
Affiliation(s)
- Xinyu Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Jinfeng Zhao
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Simin Wu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Peng Liao
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China; Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, 410219, China.
| | - Guiping Guan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China.
| |
Collapse
|
5
|
Chung KS, Heo SW, Lee JH, Han HS, Kim GH, Kim YR, Kim MS, Hong JE, Rhee KJ, Lee KT. Protective potential of nodakenin in high-fat diet-mediated colitis-associated cancer: Inhibition of STAT3 activation and Wnt/β-catenin pathway, and gut microbiota modulation. Int Immunopharmacol 2025; 157:114734. [PMID: 40318275 DOI: 10.1016/j.intimp.2025.114734] [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: 12/23/2024] [Revised: 04/14/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
A high-fat diet (HFD) exerts complex effects on the risk of colitis-associated cancer (CAC). Nodakenin, a key phytochemical isolated from the dried roots of Angelicae gigas Nakai (Umbelliferae), possesses anti-inflammatory and anti-adipogenic properties and shows potential as a therapeutic agent for colorectal cancer (CRC). In this study, we investigated the protective effects and underlying molecular mechanisms of nodakenin in an animal model of CRC induced by HFD, azoxymethane (AOM), and dextran sodium sulfate (DSS). Oral administration of nodakenin significantly alleviated clinical symptoms, such as recovery of weight, spleen weight, and colon length, and suppressed tumor progression in the colonic tissues of HFD/AOM/DSS-induced CRC mice. Nodakenin inhibited the activation of STAT3-related inflammatory mediators and downregulated proteins involved in the Wnt/β-catenin signaling pathway. These effects contributed to the disruption of epithelial-mesenchymal transition (EMT) and the restoration of tight junction integrity within the colonic tissue. Furthermore, nodakenin treatment improved the composition of the gut microbiota, leading to observable species-level differences. Network analysis revealed significant correlations between clinical parameters, inflammatory markers, EMT and apoptotic factors, and the composition of the gut microbiota. Specifically, negative correlations were observed between spleen weight and Alistipes, as well as between MCP-1 and Clostridium_g21. Positive correlations with spleen weight were observed with species belonging to Anaerotruncus, Emergencia, and Parvibacter. Bacteroidaceae_uc and Bacteroides correlated positively with MCP-1, Streptococcus correlated positively with PUMA, and Harryflintia, Odoribacteraceae_uc, and Roseburia correlated positively with cleaved caspase-3. Overall, our findings suggested that nodakenin effectively alleviates HFD/AOM/DSS-induced CRC by targeting inflammatory pathways (STAT3 and Wnt/β-catenin), suppressing EMT, and restoring gut microbiota balance. These multiple mechanisms underscore its potential as a promising agent for the prevention and treatment of colitis-associated colorectal cancer.
Collapse
Affiliation(s)
- Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - So-Won Heo
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jung-Hun Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hee-Soo Han
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gi-Hui Kim
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ye-Rin Kim
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Min-Su Kim
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
6
|
Zhang J, Guo H, Gong C, Shen J, Jiang G, Liu J, Liang T, Guo L. Therapeutic targets in the Wnt signaling pathway: Treating cancer with specificity. Biochem Pharmacol 2025; 236:116848. [PMID: 40049295 DOI: 10.1016/j.bcp.2025.116848] [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: 12/16/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 04/20/2025]
Abstract
The Wnt signaling pathway is a critical regulatory mechanism that governs cell cycle progression, apoptosis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, and the tumor immune microenvironment, while also maintaining tissue homeostasis. Dysregulated activation of this pathway is implicated in various cancers, closely linked to tumor initiation, progression, and metastasis. The Wnt/β-catenin axis plays a central role in the pathogenesis of common cancers, including colorectal cancer (CRC), breast cancer (BC), liver cancer, and lung cancer. Unlike traditional chemotherapy, targeted therapy offers a more precise approach to cancer treatment. As a key regulator of oncogenesis, the Wnt pathway represents a promising target for clinical interventions. This review provides a comprehensive analysis of the Wnt signaling pathway, exploring its roles in tumor biology and its implications in human malignancies. It further examines the molecular mechanisms and modes of action across different cancers, detailing how the Wnt pathway contributes to tumor progression through mechanisms such as metastasis promotion, immune modulation, drug resistance, and enhanced cellular proliferation. Finally, therapeutic strategies targeting Wnt pathway components are discussed, including inhibitors targeting extracellular members, as well as those within the cell membrane, cytoplasm, and nucleus. The potential of these targets in the development of novel therapeutic agents underscores the critical importance of intervening in the Wnt signaling pathway for effective cancer treatment.
Collapse
Affiliation(s)
- Jiaxi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Chengxuan Gong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jie Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Guijie Jiang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jiarui Liu
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China.
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
7
|
Jia H, Bian C, Chang Y. Exploring the molecular interactions between ferroptosis and the Wnt/β-catenin signaling pathway: Implications for cancer and disease therapy. Crit Rev Oncol Hematol 2025; 210:104674. [PMID: 40010619 DOI: 10.1016/j.critrevonc.2025.104674] [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: 01/02/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025] Open
Abstract
Ferroptosis, a regulated form of cell death dependent on iron and marked by lipid peroxidation, is increasingly recognized for its role in a wide array of diseases, including cancers, neurodegenerative disorders, and tissue damage. This review examines the dynamic interaction between ferroptosis and the Wnt/β-catenin signaling pathway, focusing on how Wnt surface receptors, ligands, antagonists, and associated components influence the regulation of ferroptosis. Key elements such as Frizzled receptors, Wnt ligands, and antagonists like DKK1 are shown to affect ferroptosis by altering oxidative stress, lipid dynamics, and iron metabolism. A central aspect of this interaction is the role of the destruction complex, particularly GSK-3β, which regulates ferroptosis through its upstream modulation by the AKT pathway and downstream control over NRF2, GPX4, and SLC7A11. Furthermore, the involvement of β-catenin/TCF transcription factors in the regulation of ferroptosis emphasizes the significance of this pathway in promoting cell survival and resisting ferroptosis, particularly in various cancers. Multiple cancers, including colorectal, breast, ovarian, and lung cancers, are affected by disruptions in the Wnt/ferroptosis axis, where enhanced Wnt signaling helps cancer cells evade ferroptosis and develop resistance to treatments. Beyond cancer, this axis also plays a crucial role in neurodegenerative diseases and conditions like myocardial infarction. Additionally, natural compounds have shown potential in modulating the Wnt/ferroptosis pathway, offering promising therapeutic approaches for a variety of diseases. This review highlights the molecular mechanisms of the Wnt/ferroptosis axis, paving the way for innovative treatment options in cancer and other diseases.
Collapse
Affiliation(s)
- Hui Jia
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Che Bian
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China.
| | - Yi Chang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
8
|
Lin T, Li Z, Yuan J, Ren T, Pang W, Xu S. Design, synthesis and evaluation of diphenyl ether-based kaiso inhibitors with enhanced potency. Bioorg Med Chem Lett 2025; 121:130158. [PMID: 40049243 DOI: 10.1016/j.bmcl.2025.130158] [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: 12/12/2024] [Revised: 02/10/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025]
Abstract
Kaiso, a potential target for the treatment of lung cancer. Our research focuses on Kaiso inhibitros. Through virtual screening and molecular dynamic simulations, we discovered a promising Kaiso inhibitor called compound 5 (ZINC20577650). By modifying the structure of compound 5, a series of novel Kaiso inhibitors that contain a diphenyl ether ring were synthesized. Among them, compound 20 exhibited the strongest inhibitory activity against A549 cells (IC50 = 0.34 μM). Notably, its inhibitory activity surpassed that of the positive control MIRA-1 (IC50 = 654.065 μM). Molecular docking and dynamic studies revealed that the binding of the compound's amino and ester moieties to the active site of kaiso protein, as well as the extension of the benzene ring towards the Asn561 position in the cavity, contributed significantly to its potency. These findings provide valuable insights for the development of new Kaiso inhibitors.
Collapse
Affiliation(s)
- Taofeng Lin
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhongqi Li
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang, China
| | - Juanchan Yuan
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Tinfeng Ren
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wan Pang
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Songhui Xu
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang, China; Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China.
| |
Collapse
|
9
|
Kahkesh S, Hedayati N, Rahimzadeh P, Farahani N, Khoozani MF, Abedi M, Nabavi N, Naeimi B, Khoshnazar SM, Alimohammadi M, Alaei E, Mahmoodieh B. The function of circular RNAs in regulating Wnt/β-catenin signaling: An innovative therapeutic strategy for breast and gynecological cancers. Pathol Res Pract 2025; 270:155944. [PMID: 40228402 DOI: 10.1016/j.prp.2025.155944] [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: 01/07/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
Breast cancer (BC) and gynecological malignancies, including cervical, ovarian, and uterine cancers, are significant global health challenges due to their high prevalence, complex nature, and elevated mortality rates. Dysregulation of the Wnt/β-catenin signaling pathway is a common feature in gynecological malignancies, contributing to cancer cell growth, progression, migration, and metastasis. Recent studies have highlighted the pivotal role of non-coding RNAs (ncRNAs), particularly circular RNAs (circRNAs), in modulating the Wnt/β-catenin signaling pathway. Acting as sponges for microRNAs (miRNAs), circRNAs regulate key oncogenic and tumor-suppressive processes by influencing Wnt-related components. This research explores the role of circRNAs in breast and gynecological malignancies, focusing on their regulatory effects on the Wnt/β-catenin pathway. The findings reveal that circRNAs modulate critical cellular processes such as proliferation, apoptosis, autophagy, and metastasis, with potential implications for therapeutic interventions. Targeting circRNA-mediated dysregulation of Wnt signaling could offer novel strategies for improving diagnostic precision, treatment efficacy, and survival outcomes in breast and gynecological cancers.
Collapse
Affiliation(s)
- Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Farhadi Khoozani
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Bita Naeimi
- Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
10
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [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: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
11
|
Yang MH, Basappa B, Deveshegowda SN, Ravish A, Mohan A, Nagaraja O, Madegowda M, Rangappa KS, Deivasigamani A, Pandey V, Lobie PE, Hui KM, Sethi G, Ahn KS. A novel drug prejudice scaffold-imidazopyridine-conjugate can promote cell death in a colorectal cancer model by binding to β-catenin and suppressing the Wnt signaling pathway. J Adv Res 2025; 72:615-632. [PMID: 39067696 DOI: 10.1016/j.jare.2024.07.022] [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: 03/28/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
INTRODUCTION Globally, colorectal cancer (CRC) is the third most common type of cancer, and its treatment frequently includes the utilization of drugs based on antibodies and small molecules. The development of CRC has been linked to various signaling pathways, with the Wnt/β-catenin pathway identified as a key target for intervention. OBJECTIVES We have explored the impact of imidazopyridine-tethered chalcone-C (CHL-C) in CRC models. METHODS To determine the influence of CHL-C on apoptosis and autophagy, Western blot analysis, annexin V assay, cell cycle analysis, acridine orange staining, and immunocytochemistry were performed. Next, the activation of the Wnt/β-catenin signaling pathway and the anti-cancer effects of CHL-C in vivo were examined in an orthotopic HCT-116 mouse model. RESULTS We describe the synthesis and biological assessment of the CHL series as inhibitors of the viability of HCT-116, SW480, HT-29, HCT-15, and SNU-C2A CRC cell lines. Further biological evaluations showed that CHL-C induced apoptosis and autophagy in down-regulated β-catenin, Wnt3a, FZD-1, Axin-1, and p-GSK-3β (Ser9), and up-regulated p-GSK3β (Tyr216) and β-TrCP. In-depth analysis using structure-based bioinformatics showed that CHL-C strongly binds to β-catenin, with a binding affinity comparable to that of ICG-001, a well-known β-catenin inhibitor. Additionally, our in vivo research showed that CHL-C markedly inhibited tumor growth and triggered the activation of both apoptosis and autophagy in tumor tissues. CONCLUSION CHL-C is capable of inducing apoptosis and autophagy by influencing the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Min Hee Yang
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Suresha N Deveshegowda
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Akshay Ravish
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Arunkumar Mohan
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Omantheswara Nagaraja
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Mahendra Madegowda
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Kanchugarakoppal S Rangappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, 169610, Singapore
| | - Vijay Pandey
- Shenzhen Bay Laboratory, Shenzhen 518055, China; Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Peter E Lobie
- Shenzhen Bay Laboratory, Shenzhen 518055, China; Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, 169610, Singapore.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Liu S, Li Q, Niu L, Feng P, Li W, Dang Y, Jia J, Yuan G, Pan Y. KCNE3 Facilitates M1 Macrophage Polarization by Suppressing the Wnt/β-Catenin Pathway, Inhibiting Glioma Proliferation, Migration, and Invasion. Mol Carcinog 2025; 64:1090-1103. [PMID: 40146943 DOI: 10.1002/mc.23911] [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: 09/29/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
The glioma microenvironment is critical for tumor growth, where reprogramming M2-polarized tumor-associated macrophages/microglia (TAMs) to an antitumor M1 phenotype represents a promising therapeutic strategy. While the potassium channel regulatory subunit KCNE3 has been implicated in tumorigenesis across malignancies, its functional role in shaping the glioma microenvironment remains undefined. Here, we leveraged transcriptome data from the Gene Expression Omnibus (GEO) to identify KCNE3 as a TAM-enriched gene in gliomas. To interrogate its mechanistic contributions, we generated KCNE3-knockdown and overexpressing macrophages and evaluated their impact on glioma cells in coculture systems. Silencing KCNE3 in macrophages significantly attenuated glioma cell proliferation, migration, and invasion in vitro, accompanied by enhanced M1 polarization. Mechanistically, KCNE3 depletion suppressed Wnt/β-catenin signaling, driving increased secretion of pro-inflammatory cytokines TNF-α, IL-6, and IL-12. Conversely, KCNE3 overexpression reversed these effects, promoting M2-like characteristics and tumor-supportive behaviors. These findings establish KCNE3 as a key modulator of TAM phenotype and glioma progression, suggesting that targeted KCNE3 inhibition may disarm pro-tumorigenic immune responses to improve therapeutic outcomes. This study uncovers a novel actionable method in glioma immunotherapy.
Collapse
Grants
- The study is supported by the National Natural Science Foundation of China (Grant Numbers: 81960541 and 82060455); the Natural Science Foundation of Gansu Province (Grant Numbers: 2021RCXM096, 21JR7RA420, 21JR7RA426, 21JR7RA411, 22JR11RA079, 22ZD6F A021-4, 22JR5RA966, 22JR5R959), the Lanzhou Science and Technology Bureau Project (Grant Numbers: 2021-RC-97), Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital (Grant Numbers: CY2022-QN-A12, CY2022-QN-B05, CY2021-MS-A09, CY2021-MS-A19, CY2021-QN-B03).
Collapse
Affiliation(s)
- Shangyu Liu
- The Second Medical College of Lanzhou University, Lanzhou, China
| | - Qiao Li
- The Second Medical College of Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| | - Liang Niu
- The Second Medical College of Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| | - Peng Feng
- The Second Medical College of Lanzhou University, Lanzhou, China
| | - Wenshan Li
- The Second Medical College of Lanzhou University, Lanzhou, China
| | - Ying Dang
- The Second Medical College of Lanzhou University, Lanzhou, China
| | - Juan Jia
- The Second Medical College of Lanzhou University, Lanzhou, China
- Department of Anesthesiology, Second Hospital of Lanzhou University, Lanzhou, China
| | - Guoqiang Yuan
- The Second Medical College of Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| | - Yawen Pan
- The Second Medical College of Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
13
|
Huo J, Nie K, Yang T, Zhang S, Zhu Z, Peng X, Zhang Y. Network pharmacology combined with transcriptomics reveals that Ganoderma lucidum spore and Sanghuangporus vaninii compound extract exerts anti-colorectal cancer effects via CYP24A1-mediated VDR pathway and TERT-mediated Wnt signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119820. [PMID: 40245966 DOI: 10.1016/j.jep.2025.119820] [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: 01/12/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As traditional medicinal fungi, Ganoderma lucidum and Sanghuangporus vaninii are widely used in the treatment of tumor related diseases and cancer adjuvant therapy with potent anticancer effects. However, the anticancer effect and mechanism of action of their compound extract remain unclear. AIM OF THE STUDY To investigate the anticancer effect of Ganoderma lucidum and Sanghuangporus vaninii compound extract and explore the underlying mechanism. MATERIALS AND METHODS First, MTT assay was performed to investigate the effect of 8 different extracts on tumor cell viability. Moreover, the synergistic effect of Ganoderma lucidum spore and Sanghuangporus vaninii was evaluated by Chou-Talalay method. Subsequently, the fractional extractions were conducted to further isolate anti-tumor active components. Next, network pharmacology combined with transcriptomics was used to explore the potential mechanisms underlying the anticancer effect of compound extract. Finally, the mechanism of action was verified using in vitro and in vivo models. RESULTS Among all 8 extracts, Ganoderma lucidum spore and Sanghuangporus vaninii compound ethanol extract (GSEE) showed the most significant cell viability inhibitory effect on cancer cells, especially colorectal cancer (CRC) cells, which was even better than combination of Sanghuangporus vaninii ethanol extract (SVEE) and Ganoderma lucidum spore ethanol extract (GLEE). The ethyl acetate fraction of GSEE (GSEAE) was screened as the anti-tumor active fraction of GSEE and could suppress CRC proliferation in vitro and in vivo. The CYP24A1-mediated Vitamin D receptor (VDR) pathway and TERT-mediated Wnt signaling pathway were identified as the main mechanisms of GSEAE against CRC. Multiple CRC models confirmed that GSEAE suppressed CRC metastasis, arrested cell cycle and induced mitochondrial apoptosis of CRC cells via VDR pathway and Wnt signaling pathway. CONCLUSIONS Collectively, our data suggest that compound extract GSEAE exerts anti-CRC effects via CYP24A1-mediated VDR pathway and TERT-mediated Wnt signaling pathway.
Collapse
Affiliation(s)
- Jian Huo
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Kun Nie
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Tianfeng Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Suyu Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Xiuhong Peng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an, 710061, PR China.
| |
Collapse
|
14
|
Li Y, Liu Y, Wu A, Liu H, Liang M, Pan Q, Cheng D. Aptamer inhibits P-glycoprotein efflux function via the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2025; 760:151709. [PMID: 40168709 DOI: 10.1016/j.bbrc.2025.151709] [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: 01/20/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Inhibiting permeability glycoprotein (P-gp) efflux is a strategy to enhance drug efficacy or overcome multidrug resistance in tumors. However, whether P-gp aptamer (APTP-gp, an 81 bp ssDNA) inhibits P-gp efflux is unknown. Increased Rho123 uptake was observed in the rat brain and intestine. Bidirectional transport of Rho123 indicated that 100 nM of APTP-gp inhibited P-gp activity with inhibition ratios of 75.0 % in Caco-2 and 60.5 % in hCMEC/D3 cells. The apparent permeability coefficients (Papp) from the apical (AP) to basolateral (BL) sides significantly increased by 129.4 % in Caco-2 and 8.0 % in hCMEC/D3 cells, respectively. The Papp from the BL→AP sides in the two cell lines decreased. P-gp mRNA and protein expression in the rat ileum, brain, and two cell lines markedly decreased following APTP-gp exposure. APTP-gp downregulated Wnt3, pho-Dvl2, β-catenin expression and decreased the ratio of pho-GSK-3β to GSK-3β in the rat ileum and brain. Molecular docking analysis suggested that APTP-gp interact with Wnt/β-catenin signaling pathway proteins at various amino acid sites. The present study reports a novel a novel nucleic acid-based P-gp inhibitor, which may benefit for enhancing drug efficacy or overcome multidrug resistance in clinical application.
Collapse
Affiliation(s)
- Yujuan Li
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Yujiao Liu
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Aijia Wu
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Huayan Liu
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Min Liang
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Qiuxia Pan
- People's Liberation Army Strategic Support Force Characteristic Medical Center, Beijing, 100101, China.
| | - Dongsheng Cheng
- Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China.
| |
Collapse
|
15
|
Wu D, Xia Q, Su X, Mao Y, Mao J, Ding Q, Liu J, Zhong W, Zhang X, Li H, Duan S. Long non-coding RNA TMEM51-AS1 inhibits colorectal cancer progression. Discov Oncol 2025; 16:878. [PMID: 40407985 PMCID: PMC12102048 DOI: 10.1007/s12672-025-02676-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/12/2025] [Indexed: 05/26/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cause of death worldwide and has high mortality and a poor prognosis. Long non-coding RNAs (lncRNAs) are non-coding RNAs longer than 200 nucleotides that play roles in cancer through multiple mechanisms. TMEM51-AS1 is a newly discovered 40,650 bp lncRNA. Our results showed that TMEM51-AS1 expression was significantly downregulated in CRC tissues (fold change = 0.74, P < 0.0001). This finding was confirmed in 20 pairs of CRC carcinoma and paracancerous tissues (fold change = 0.5, P < 0.001). Additionally, TMEM51-AS1 expression was found to be significantly reduced in CRC cell lines compared to normal human intestinal epithelial cells (P < 0.001). Bioinformatic analysis revealed that TMEM51-AS1 expression was associated with immune escape, RNA methylation, and DNA damage and repair. TMEM51-AS1 may also activate energy metabolism pathways to participate in cancer development. Drug sensitivity analysis confirmed that several drugs are more effective in CRC patients with high expression of TMEM51-AS1. In conclusion, our study demonstrates that TMEM51-AS1 can suppress the progression of CRC.
Collapse
Affiliation(s)
- Dongping Wu
- Department of Radiation Oncology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Qing Xia
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310014, Zhejiang, China
| | - Xinming Su
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310014, Zhejiang, China
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Yunan Mao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310014, Zhejiang, China
| | - Jiwei Mao
- Department of Radiation Oncology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Qiannan Ding
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Jianjiang Liu
- Department of Radiation Oncology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Wangyan Zhong
- Department of Radiation Oncology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Xiaoyu Zhang
- Department of Radiation Oncology, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Hanbing Li
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China.
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
16
|
An C, Jiang C, Pei W, Li A, Wang M, Wang Y, Wang H, Zuo L. Intestinal epithelial cells in health and disease. Tissue Barriers 2025:2504744. [PMID: 40401816 DOI: 10.1080/21688370.2025.2504744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
This comprehensive review delves into the pivotal role of intestinal epithelial cells in the context of various diseases. It provides an in-depth analysis of the diverse types and functions of these cells, explores the influence of multiple signaling pathways on their differentiation, and elucidates their critical roles in a spectrum of diseases. The significance of the gastrointestinal tract in maintaining overall health is extremely important and cannot be exaggerated. This complex and elongated organ acts as a crucial link between the internal and external environments, making it vulnerable to various harmful influences. Preserving the normal structure and function of the gut is essential for well-being. Intestinal epithelial cells serve as the primary defense mechanism within the gastrointestinal tract and play a crucial role in preventing harmful substances from infiltrating the body. As the main components of the digestive system, they not only participate in the absorption and secretion of nutrients and the maintenance of barrier function but also play a pivotal role in immune defense. Therefore, the health of intestinal epithelial cells is of vital importance for overall health.
Collapse
Affiliation(s)
- Chenchen An
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| | - Chonggui Jiang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| | - Wangxiang Pei
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| | - Ao Li
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
- The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, China
| | - Minghui Wang
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yufei Wang
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Hua Wang
- Inflammation and Immune- Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Zuo
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for college students, Anhui Medical University, Hefei, China
| |
Collapse
|
17
|
Zeng Z, Cai S, Ye C, Li T, Tian Y, Liu E, Cai J, Yuan X, Yang H, Liang Q, Li K, Peng C. Neural influences in colorectal cancer progression and therapeutic strategies. Int J Colorectal Dis 2025; 40:120. [PMID: 40379990 PMCID: PMC12084286 DOI: 10.1007/s00384-025-04887-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2025] [Indexed: 05/19/2025]
Abstract
PURPOSE This review aims to elucidate the neural mechanisms driving colorectal cancer (CRC) growth, metastasis, and therapeutic resistance, summarizing the roles of neurotransmitters, neurotrophic factors, and neural signaling in carcinogenesis. It further explores therapeutic strategies targeting neural dependencies in CRC. METHODS A comprehensive PubMed search was conducted using the keywords colorectal cancer and tumor innervation, focusing on studies published between 2000 and 2024. The review synthesizes evidence across four domains: neurotransmitter-receptor interactions, gut-brain-microbiota axis dynamics, neuroimmune modulation, and neural regulation of cancer stem cells, discussing their collective impact on CRC pathophysiology. RESULTS Neural innervation significantly influences CRC progression. For instance, the neurotransmitter serotonin promotes tumor growth and metastasis via paracrine and autocrine stimulation, while neurotrophic mediators like nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) activate oncogenic signaling through receptor tyrosine kinases (RTKs). Downstream pathways, such as Wnt/β-catenin signaling, are modulated by neural inputs, underscoring CRC's neurodevelopmental dependency and highlighting their potential as therapeutic targets. CONCLUSION Neural mechanisms are pivotal in CRC progression, revealing novel therapeutic avenues. Strategies targeting neurotransmitter synthesis, neurotrophic signaling, or neuroimmune crosstalk may disrupt tumorigenic loops while preserving systemic nervous system integrity. Future research must prioritize translating these insights into clinical interventions to improve patient outcomes. Elucidating the intricate interplay between neural mediators and cancer pathogenesis, coupled with developing therapies specifically targeting the neurogenic basis of CRC aggressiveness, represents a critical frontier in oncology.
Collapse
Affiliation(s)
- Zhibin Zeng
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Shirong Cai
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Chenle Ye
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Tongduan Li
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Yan Tian
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Enyuan Liu
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Junbin Cai
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Xiaojun Yuan
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Heng Yang
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Quanqi Liang
- Division of Gastroenterology, Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China
| | - Kaishu Li
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China.
| | - Cui Peng
- Department of Gynaecology and Obstetrics, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, China.
| |
Collapse
|
18
|
Zhang J, Liu Z, Ma X, Shi Z, Zhao J, Xie Y, Shang X, Zhang X. Deciphering the interaction between the expression of LRP2 served as a mitochondrial metabolism-related gene and prognosis in colon cancer integrating multi-omics analysis. Discov Oncol 2025; 16:782. [PMID: 40377809 PMCID: PMC12084449 DOI: 10.1007/s12672-025-02568-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 05/05/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Colon adenocarcinoma (COAD) is increasingly prevalent among patients under 50 years old, and the 5-year survival rate for patients with metastasis is less than 20%. Identifying significant biomarkers and therapeutic targets is crucial. We investigated the expression of LRP2 in COAD and its prognostic value utilizing single-cell sequencing and transcriptomics datasets, which was conducted preliminary validation at the patient samples and cellular levels as well. METHODS Based on differential gene expression of tumor samples and normal tissues in The Cancer Genome Atlas (TCGA), we performed consensus clustering, univariate and multivariate Cox regression analysis applying 1,234 mitochondrial metabolism-related genes (MMRGs) to identify some essential genes associated with poor prognosis in COAD patients. We validated survival outcome and biological function of the target gene leveraging single-cell sequencing and transcriptomics datasets from Gene Expression Omnibus (GEO), and evaluated the value of the target gene in the clinical pathology stage of COAD patients. Simultaneously, the expression levels of critical gene were detected in the diverse tissues of COAD by immunohistochemistry (IHC) staining. Transcriptomics data was continuously implemented to compare the discrepancy between the expression levels of the target gene and somatic mutation burden, inspecting the key pathways of the target gene by gene set enrichment analysis (GSEA) and examining its drug sensitivity synthetically in the CellMiner databases. The proliferative capacity augmented in LRP2-overexpressed colon cancer cells was determined employing cell counting kit-8 (CCK-8) and flow cytometry assays. RESULTS LRP2 served as a key mitochondrial metabolism-related gene was assessed clinical prognosis in COAD patients according to the TCGA database. High expression of LRP2 was prominently associated with poor prognosis in COAD patients (P < 0.05), which was validated by GEO databases, and the expression levels of LRP2 were positively related to clinical pathological stage simultaneously (P < 0.05). Some specific cell types were clustered and proliferation pathways were immensely enriched, which were correlated with LRP2 in two single-cell sequencing datasets. The mutation profiles displayed remarkable differences in two levels of LRP2, we also observed high expressions of LRP2 were immensely correlated with high tumor mutation burden (TMB) and unfavorable prognosis in these patients (P < 0.05). LRP2 was significantly enriched in multiple cancer proliferation-related pathways, and the noteworthy correlation between LRP2 and the sensitivity to various drugs was identified (P < 0.05). The expression levels of LRP2 were multifarious in different COAD patients based on IHC staining. LRP2-overpression could stimulate the proliferation capability of HCT116 and SW480 cell lines markedly (P < 0.05). CONCLUSION The expression levels of LRP2 were intimately correlated with gene mutations, prognosis, pathological stage and the sensitivity to anticancer drugs in COAD. Augmented levels of LRP2 would manifest poor prognosis, which furnished novel insights for clinical diagnosis and treatment in COAD. LRP2 could extensively facilitate the proliferation ability of colon cell lines.
Collapse
Affiliation(s)
- Jie Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Ziyun Liu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xiaoqing Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Zhenyu Shi
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jing Zhao
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yongjie Xie
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Xiaobin Shang
- Department of Minimally Invasive Esophageal Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xia Zhang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
19
|
Li YW, Liu Y, Luo SZ, Huang XJ, Shen Y, Wang WS, Lang ZC. The significance of calcium ions in cerebral ischemia-reperfusion injury: mechanisms and intervention strategies. Front Mol Biosci 2025; 12:1585758. [PMID: 40421420 PMCID: PMC12104078 DOI: 10.3389/fmolb.2025.1585758] [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: 03/01/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Cerebral ischemia-reperfusion injury (CIRI) represents a multifaceted pathological phenomenon characterized by an array of molecular and cellular mechanisms, which significantly contribute to neurological dysfunction. Evidence suggests that calcium ions play an indispensable role in this context, as abnormal elevations in calcium concentrations exacerbate neuronal injury and intensify functional deficits. These ions are integral not only for intracellular signaling pathways but also for various pathological processes, such as programmed cell death, inflammatory responses, and oxidative stress. This review article elucidates the physiological framework of calcium homeostasis and the precise mechanisms through which calcium ions influence CIRI. Moreover, it addresses potential intervention strategies, including calcium channel blockers, calmodulin (CaM) inhibitors, antioxidants, and anti-inflammatory agents. Despite the proposal of certain intervention strategies, their effectiveness and safety in clinical settings warrant further scrutiny. In conclusion, the article highlights the limitations of current research and anticipates future investigative trajectories, aiming to provide a theoretical foundation and reference for the development of more efficacious treatment modalities.
Collapse
|
20
|
Chen S, Jiang Z, Song W, Lu C, Lin Y, Xu S, Xie K, Wan L, Yuan X. Identification of the "Collagen-Macrophage" sub-category of patients with colorectal cancer as an extension of the CMS4 subtype with THBS2 as a therapeutic target. BMC Gastroenterol 2025; 25:342. [PMID: 40340827 PMCID: PMC12060322 DOI: 10.1186/s12876-025-03918-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
We identified a subset of patients with colorectal cancer (CRC) enriched with "collagen-TAMs," designated the CM class, using large integrated colon cancer transcriptome and single-cell transcriptome datasets. This classification system could be used as an extension of the traditional CMS classification system for CRC to guide more accurate classification and treatment.We also screened CAF-derived THBS2 as a potential biomarker for CM and found that it plays an important role in CRC disease models in vitro and in vivo, promoting tumor development and metastasis as well as TAM recruitment. Targeting THBS2 combined with PD-1 therapy effectively improved the therapeutic effect of immunotherapy in vivo. The CM classification provides a new perspective for CRC treatment, and THBS2, which is highly expressed in CM cases, can be used as a new potential combined target for immunotherapy.
Collapse
Affiliation(s)
- Shuwen Chen
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, 211166, China
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Zhaoyan Jiang
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, 211166, China
| | - Wanxuan Song
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Chuqiao Lu
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Yanbing Lin
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Shiyao Xu
- Department of Clinical Medicine, First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China
| | - Kunxin Xie
- Department of Biochemistry and Molecular Biology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| | - Li Wan
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223302, China.
| | - Xiaoqin Yuan
- Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
21
|
Wang P, Li G, Sun X, Zhang J, Shi L, Zhou X, Wang G, Chen W. miR-182-5p facilitates colorectal cancer progression through manipulating neurocalcin delta mediated Wnt/β-catenin signalling. Eur J Med Res 2025; 30:352. [PMID: 40312722 PMCID: PMC12046800 DOI: 10.1186/s40001-025-02625-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/22/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC), a complex and multifactorial disease, has been associated with elevated expression of microRNA miR-182-5p, although its precise regulatory role in CRC progression remains unclear. This study aims to identify potential therapeutic targets to improve clinical outcomes and to decipher the intricate role of miR-182-5p in the pathobiology of CRC. METHODS We conducted comprehensive bioinformatics analyses using GEO databases to investigate differences in miRNA expression between CRC and normal tissues, with a particular focus on miR-182-5p. Its expression levels in CRC cells and tumor tissues were quantified by quantitative real-time PCR (qRT-PCR). The expression of neurocalcin delta (NCALD) and proteins related to Wnt/β-catenin signalling was evaluated by qRT-PCR and Western blotting. Pathological changes in tumor-bearing mice as well as the proliferation, invasion, and migration of CRC cells, were assessed. Tumor cell proliferation and apoptosis were examined using Ki-67 immunohistochemistry and TUNEL staining, respectively. A dual luciferase reporter assay explored the regulatory interaction between miR-182-5p and NCALD. RESULTS Our findings reveal significantly elevated miR-182-5p levels in CRC tissues and cell lines, positively correlated with tumor invasion depth, differentiation degree, clinical stage, and lymph node metastasis. miR-182-5p appears to accelerate CRC progression in both cell lines and mouse models by downregulating NCALD, thereby enhancing Wnt/β-catenin signalling. This study identifies miR-182-5p as a pivotal enhancer of CRC progression, modulating Wnt/β-catenin signalling via NCALD regulation. CONCLUSIONS The findings position the miR-182-5p/NCALD axis as promising targets for CRC therapy, offering new avenues for treatment strategies. TRIAL REGISTRATION Retrospectively registered.
Collapse
Affiliation(s)
- Pengfei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Department of Gastroenterology, Affiliated Qidong Hospital of Nantong University, Qidong, 226200, Jiangsu, China
| | - Gang Li
- Institute of Special Environmental Medicine, Nantong University, Chongchuan District, 9 Seyuan Road, Nantong, 226019, Jiangsu, China
| | - Xianglin Sun
- Institute of Special Environmental Medicine, Nantong University, Chongchuan District, 9 Seyuan Road, Nantong, 226019, Jiangsu, China
| | - Jie Zhang
- Department of Gastroenterology, Affiliated Qidong Hospital of Nantong University, Qidong, 226200, Jiangsu, China
| | - Leijian Shi
- Department of Gastroenterology, Affiliated Qidong Hospital of Nantong University, Qidong, 226200, Jiangsu, China
| | - Xiaoyu Zhou
- Department of Gastroenterology, Affiliated Qidong Hospital of Nantong University, Qidong, 226200, Jiangsu, China
| | - Guohua Wang
- Institute of Special Environmental Medicine, Nantong University, Chongchuan District, 9 Seyuan Road, Nantong, 226019, Jiangsu, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
22
|
Lee J, Kim I, Ryu J, Eling T, Baek SJ. NAG-1/GDF15 as a tumor suppressor in colorectal cancer: inhibition of β-catenin and NF-κB pathways via interaction with EpCAM. Cell Death Dis 2025; 16:355. [PMID: 40316530 PMCID: PMC12048721 DOI: 10.1038/s41419-025-07695-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
NAG-1/GDF15, a tumor suppressor, is synthesized as a pro-form in colorectal cancer (CRC) cells and undergoes cleavage to generate its mature form. While the biological function of pro-NAG-1/GDF15 remains unclear, our study reveals its crucial role in suppressing oncogenic signaling. We demonstrate that pro-NAG-1/GDF15 is predominantly retained within cells, whereas its mature form is secreted into the media. The expression of NAG-1/GDF15, or uncleavable R193A mutant, inhibits β-catenin and NF-κB signaling, key pathways in CRC progression. Mechanistically, the pro-NAG-1/GDF15 interacts with EpCAM, preventing its cleavage and nuclear translocation, thereby reducing β-catenin and NF-κB activity. This inhibition correlates with decreased expression of oncogenic targets such as cyclin D1 and c-myc. In vivo, NAG-1/GDF15 expression significantly reduces tumor growth in cancer xenograft models, accompanied by decreased proliferation and increased apoptosis. Furthermore, analysis of public datasets suggests that high NAG-1/GDF15 expression is associated with improved CRC patient survival. These findings highlight NAG-1/GDF15 via the formation of pro-NAG-1/GDF15 as a promising therapeutic target for cancer, with potential applications in modulating tumorigenic signaling pathways.
Collapse
Affiliation(s)
- Jaehak Lee
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Ilju Kim
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Junsun Ryu
- Department of Otolaryngology-Head and Neck Surgery, Center for Thyroid Cancer, Research Institute and Hospital, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Thomas Eling
- Retired Scientist Emeritus, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Seung Joon Baek
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea.
| |
Collapse
|
23
|
Sun MX, Zhu HC, Yu Y, Yao Y, Li HY, Feng FB, Wang QY, Liu RJ, Sun CG. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int J Oncol 2025; 66:36. [PMID: 40145557 PMCID: PMC12068849 DOI: 10.3892/ijo.2025.5742] [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: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The focus on breast cancer treatment has shifted from the cytotoxic effects of single drugs on tumor cells to multidimensional multi‑pathway synergistic intervention strategies targeting the tumor microenvironment (TME). The activation of the Wnt signaling pathway in the TME of breast cancer cells serves a key regulatory role in tissue homeostasis and is a key driver of the carcinogenic process. Modulating the crosstalk between the Wnt pathway and TME of breast cancer is key for understanding the biological behavior of breast cancer and advancing the development of novel antitumor drugs. The present review aimed to summarize the complex mechanisms of the Wnt signaling pathway in the breast cancer TME, interactions between the Wnt signaling pathway and components of the breast cancer TME and breast cancer‑associated genes, as well as the interactions between the Wnt signaling pathway and other signaling cascades at the molecular level. Furthermore, the present review aimed to highlight the unique advantages of the Wnt signaling pathway in the macro‑regulation of the TME and the current therapeutic strategies targeting the Wnt signaling pathway, their potential clinical value and future research directions in breast cancer treatment.
Collapse
Affiliation(s)
- Meng Xuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Han Ci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Hua Yao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fu Bin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Qing Yang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Rui Juan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Chang Gang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
24
|
Guo S, Ding R, Zhao Q, Wang X, Lv S, Ji XY. Recent Insights into the Roles of PEST-Containing Nuclear Protein. Mol Biotechnol 2025; 67:1800-1813. [PMID: 38762838 DOI: 10.1007/s12033-024-01188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
PEST-containing nuclear protein (PCNP), a short-lived small nuclear protein with 178 amino acids, is a nuclear protein containing two PEST sequences. PCNP is highly expressed in several malignant tumors such as cervical cancer, rectal cancer, and lung cancer. It is also associated with cell cycle regulation and the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and Wnt signaling pathways during tumor growth. The present article discuss how PCNP regulates the PI3K/AKT/mTOR and Wnt signaling pathways and related proteins, and the ubiquitination of PCNP regulates tumor cell cycle as well as the progress of the application of PCNP in the pathophysiology and treatment of colon cancer, human ovarian cancer, thyroid cancer, lung adenocarcinoma and oral squamous cell carcinoma. The main relevant articles were retrieved from PubMed, with keywords such as PEST-containing nuclear protein (PCNP), cancer (tumor), and signaling pathways as inclusion/exclusion criteria. Relevant references has been included and cited in the manuscript.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Ruidong Ding
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Qian Zhao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Xu Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Shuangyu Lv
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, Kaifeng, 475004, Henan, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Mazhai, Erqi District, Zhengzhou, 450064, Henan, China.
| |
Collapse
|
25
|
Woraharn W, McCulloch A, Bigley C, Hatthakarnkul P, Pennel K, Alexander P, van Wyk H, Roseweir A, Hay J, Maka N, Park J, Jamieson NB, Edwards J, Roxburgh CS. Investigation of three alternative histopathological scoring methods at the invasive tumour front in colorectal cancer. J Pathol Clin Res 2025; 11:e70031. [PMID: 40374531 DOI: 10.1002/2056-4538.70031] [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: 02/06/2025] [Revised: 03/27/2025] [Accepted: 04/25/2025] [Indexed: 05/17/2025]
Abstract
Although the characteristics at the invasive tumour front in colorectal cancer (CRC) are simple to assess, they are not included in routine pathology reports because they lack reproducibility and standardisation. In this study, we aimed to validate alternative scoring methods at the invasive tumour front in a large cohort of stage I-III CRC. The retrospective analysis was performed on haematoxylin and eosin-stained sections from 538 patients. At the invasive tumour front, tumour characteristics were scored using three alternative methods: the Karamitopoulou method, which evaluates the percentage of infiltrative tumour; the Taskin method, a five-point grading scale; and the tumour growth pattern (TGP) method, which classifies patterns as pushing, intermediate, or infiltrative. For interobserver assessment, the Karamitopoulou and TGP methods showed good agreement while the Taskin method presented fair agreement. High scores with the Karamitopoulou and Taskin methods correlated significantly with adverse prognostic factors, particularly advanced T stage (p < 0.001), N stage (p < 0.001), and the presence of peritoneal involvement (p < 0.001). The survival rate of the TGP method demonstrated that patients with an infiltrative growth pattern had significantly worse CRC survival compared to those with pushing and intermediate growth patterns (p < 0.001) and the TGP method retained its independence as a prognostic factor in multivariable Cox regression analysis only for colon cancer-specific survival (p < 0.001). The TGP scoring method is an independent prognostic factor only for colon cancer with simple and inexpensive assessment, underlining its practicality in routine reporting. Additionally, this method could be included as an additional histopathological risk indicator with the potential to guide therapeutic decision making.
Collapse
Affiliation(s)
| | | | | | | | - Kathryn Pennel
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Peter Alexander
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Academic Unit of Surgery, University of Glasgow, Glasgow, UK
| | - Hester van Wyk
- Academic Unit of Surgery, University of Glasgow, Glasgow, UK
| | | | - Jennifer Hay
- Glasgow Tissue Research Facility, Queen Elizabeth University Hospital, Glasgow, UK
| | - Noori Maka
- Department of Pathology, Queen Elizabeth Hospital, Glasgow, UK
| | - James Park
- Department of Surgery, Queen Elizabeth University Hospital, Glasgow, UK
| | - Nigel B Jamieson
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Academic Unit of Surgery, University of Glasgow, Glasgow, UK
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Campbell Sd Roxburgh
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Academic Unit of Surgery, University of Glasgow, Glasgow, UK
| |
Collapse
|
26
|
Rahimi A, Baghernejadan Z, Hazrati A, Malekpour K, Samimi LN, Najafi A, Falak R, Khorramdelazad H. Combination therapy with immune checkpoint inhibitors in colorectal cancer: Challenges, resistance mechanisms, and the role of microbiota. Biomed Pharmacother 2025; 186:118014. [PMID: 40157004 DOI: 10.1016/j.biopha.2025.118014] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Colorectal cancer (CRC) is still one of the leading causes of cancer deaths worldwide. Even though there has been progress in cancer immunotherapy, the results of applying immune checkpoint inhibitors (ICIs) have been unsatisfactory, especially in microsatellite stable (MSS) CRC. Single-agent ICIs that target programmed cell death-1 (PD-1)/ PD-L1, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell Ig- and mucin-domain-containing molecule-3 (TIM-3), and lymphocyte activation gene (LAG)-3 have emerged as having specific benefits. However, many primary and secondary resistance mechanisms are available in the tumor microenvironment (TME) that prevent it from happening. Combination strategies, such as the use of anti-PD-1 and anti-CTLA-4, can be effective in overcoming these resistance pathways, but toxicities remain a significant concern. Moreover, ICIs have been integrated with various treatment modalities, including chemotherapy, radiotherapy, antibiotics, virotherapy, polyadenosine diphosphate-ribose polymerase (PARP) inhibitors, and heat shock protein 90 (HSP90) inhibitors. The outcomes observed in both preclinical and clinical settings have been encouraging. Interestingly, manipulating gut microbiota via fecal microbiota transplantation (FMT) has been identified as a new strategy to increase the efficacy of immunotherapy in CRC patients. Therefore, integrating ICIs with other treatment approaches holds promise in enhancing the prognosis of CRC patients. This review focuses on the unmet need for new biomarkers to select patients for combination therapies and the ongoing work to overcome resistance and immune checkpoint blockade.
Collapse
Affiliation(s)
- Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
27
|
Xun Y, Chen G, Tang G, Zhang C, Zhou S, Fong TL, Chen Y, Xiong R, Wang N, Feng Y. Traditional Chinese medicine and natural products in management of hepatocellular carcinoma: Biological mechanisms and therapeutic potential. Pharmacol Res 2025; 215:107733. [PMID: 40209965 DOI: 10.1016/j.phrs.2025.107733] [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: 10/09/2024] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Hepatocellular carcinoma (HCC), originating from hepatocytes, is the most common type of primary liver cancer. HCC imposes a significant global health burden with high morbidity and mortality, making it a critical public concern. Surgical interventions, including hepatectomy and liver transplantation, are pivotal in achieving long-term survival for patients with HCC. Additionally, ablation therapy, endovascular interventional therapy, radiotherapy, and systemic anti-tumor therapies are commonly employed. However, these treatment modalities are often associated with considerable challenges, including high postoperative recurrence rates and adverse effects. Traditional Chinese medicine (TCM) and natural products have been utilized for centuries as a complementary approach in managing HCC and its complications, demonstrating favorable clinical outcomes. Various bioactive compounds derived from TCM and natural products have been identified and purified, and their mechanisms of action have been extensively investigated. This review aims to provide a comprehensive and up-to-date evaluation of the clinical efficacy of TCM, natural products and their active constituents in the treatment and management of HCC. Particular emphasis is placed on elucidating the potential molecular mechanisms and therapeutic targets of these agents, including their roles in inhibiting HCC cell proliferation, inducing apoptosis and pyroptosis, suppressing tumor invasion and metastasis, and restraining angiogenesis within HCC tissues.
Collapse
Affiliation(s)
- Yunqing Xun
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Guang Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Guoyi Tang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Shichen Zhou
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Tung-Leong Fong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Yue Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Ruogu Xiong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong.
| |
Collapse
|
28
|
Ghasemi N, Azizi H, Qorbanee A, Skutella T. From unipotency to pluripotency: deciphering protein networks and signaling pathways in the generation of embryonic stem-like cells from murine spermatogonial stem cells. BMC Genomics 2025; 26:426. [PMID: 40307702 PMCID: PMC12042637 DOI: 10.1186/s12864-025-11612-y] [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: 10/19/2024] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
With the significant challenges in using human embryonic stem cells (ESCs) for research and clinical applications, there is a growing impetus to seek alternative pluripotent cell sources. Embryonic stem-like (ES-like) cells emerge as a promising avenue in this pursuit. Our research demonstrates the potential for deriving ES-like cells from spermatogonial stem cells (SSCs) in a time-dependent manner under defined culture conditions. To better understand this process, we investigated the gene expression dynamics and underlying pathways associated with ES-like cell generation from SSCs. A deeper understanding of the signaling pathways underlying this biological process can lead us to refine protocols for ES-like cell generation, which could catalyze the development of more efficient and expedited methodologies inspired by the derivation pathway for future research in regenerative medicine. To identify differentially expressed genes (DEGs), we analyzed publicly available microarray data from murine cells obtained from the Gene Expression Omnibus (GEO). This analysis enabled the prediction of protein-protein interactions (PPIs), which were subsequently used for pathway enrichment analysis to identify biologically relevant pathways. Complementing these computational findings, we conducted in vitro experiments, including Fluidigm qPCR and immunostaining. These experiments serve as validation for our microarray data and the DEGs identified, providing reassurance about the reliability of our research. Among the identified enriched pathways in our investigation are the Toll-like receptor (TLR), GDNF/RET, interleukins (ILs), FGF/FGFR, and SMAD signaling pathway, along with the activation of NIMA kinases. Additionally, miR-410-3p, miRNA let-7e, Miat, and Xist are among some of the predicted non-coding RNAs.
Collapse
Affiliation(s)
- Nima Ghasemi
- Department of Applied Biotechnology and System Biology, College of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Department of Stem Cells and Cancer, College of Biotechnology, Amol University of Special Modern Technologies, P.O. Box 49767, Amol, Iran.
| | - Ali Qorbanee
- Department of Surgery, Faculty of General of Medicine, Koya University, Koya, Kurdistan Region FR, KOY45, Iraq
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Im Neuenheimer Feld 307, Heidelberg, 69120, Germany
| |
Collapse
|
29
|
Zhou S, Yu L, Zhao J, Xiao Q, Sun J, Wang L, Zhou Y, Lu Y, Dunlop MG, Theodoratou E, Zhang H, Ding K, Li X. Integration of multi-omics data to unveil the molecular landscape and role of piRNAs in early-onset colorectal cancer. BMC Med 2025; 23:250. [PMID: 40301858 PMCID: PMC12042543 DOI: 10.1186/s12916-025-04074-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/11/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND The incidence of early-onset colorectal cancer (EOCRC) (< 50 years) has been steadily rising, with a parallel increase in metastatic and invasive cases. To elucidate the molecular mechanisms underlying this aggressive phenotype, we performed comprehensive multi-omics profiling to delineate the distinct features of EOCRC, with a focus on key drivers of metastatic and invasive potential. METHODS We initially characterized the genome, epigenome, and transcriptome of tumors from 515 (69 EOCRC and 446 late-onset CRC [LOCRC]) cases in The Cancer Genome Atlas. Key candidate molecules were further validated using RNA-seq and scRNA-seq data. Multi-omics profiling revealed PIWIL1/piRNA as a hallmark of EOCRC, with further validation through in vitro functional assays, transcriptomic profiling, and Kaplan-Meier survival analysis. RESULTS EOCRC demonstrated a mutational landscape similar to that of LOCRC, with comparable oncogenic driver mutations and somatic copy-number alterations. However, EOCRC exhibited a higher frequency of deletion in chromosomes 6, 15, and 19 regions, along with metabolic reprogramming favoring aerobic glycolysis and lipid metabolism. Integrative transcriptomic and DNA methylation analyses identified six EOCRC-specific molecules, including PIWIL1. Notably, PIWIL1 was mainly expressed in epithelial cells, with lower expression in EOCRC versus LOCRC. Its downstream piRNAs (FR019019, FR019089, and FR132045) were also downregulated in EOCRC. Functional experiments demonstrated that FR019089/FR019019 overexpression suppressed migration and invasion. Clinically, low FR019089 levels correlated with significantly shorter progression-free and overall survival in EOCRC patients. Additionally, downstream pathways of FR019089 and FR019019 overexpression were enriched in anti-cancer-related signaling pathways. CONCLUSIONS Our multi-omics approach yields novel insights into the molecular underpinnings of EOCRC and we characterize the role of PIWIL1-associated piRNAs in modulating EOCRC metastasis and invasion. FR019089 shows promise as a prognostic biomarker with potential clinical utility in the risk stratification and management of EOCRC patients.
Collapse
Affiliation(s)
- Siyun Zhou
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lili Yu
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Jianhui Zhao
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Sun
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lijuan Wang
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Yuan Zhou
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yadong Lu
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Malcolm G Dunlop
- Cancer Research UK Scotland Centre and Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Scotland Centre and Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Honghe Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China.
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for CANCER, Hangzhou, China.
| | - Xue Li
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
30
|
Alhosani F, Alhamidi RS, Ilce BY, Altaie AM, Ali N, Hamad AM, Künstner A, Khandanpour C, Busch H, Al-Ramadi B, Harati R, Sayed K, AlFazari A, Bendardaf R, Hamoudi R. Transcriptome-Wide Analysis and Experimental Validation from FFPE Tissue Identifies Stage-Specific Gene Expression Profiles Differentiating Adenoma, Carcinoma In-Situ and Adenocarcinoma in Colorectal Cancer Progression. Int J Mol Sci 2025; 26:4194. [PMID: 40362431 PMCID: PMC12071244 DOI: 10.3390/ijms26094194] [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: 03/21/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Colorectal cancer (CRC) progression occurs through three stages: adenoma (pre-cancerous lesion), carcinoma in situ (CIS) and adenocarcinoma, with tumor stage playing a pivotal role in the prognosis and treatment outcomes. Despite therapeutic advancements, the lack of stage-specific biomarkers hinders the development of accurate diagnostic tools and effective therapeutic strategies. This study aims to identify stage-specific gene expression profiles and key molecular mechanisms in CRC providing insights into molecular alterations across disease progression. Our methodological approach integrates the use of absolute gene set enrichment analysis (absGSEA) on formalin-fixed paraffin-embedded (FFPE)-derived transcriptomic data, combined with large-scale clinical validation and experimental confirmation. A comparative whole transcriptomic analysis (RNA-seq) was performed on FFPE samples including adenoma (n = 10), carcinoma in situ (CIS) (n = 8) and adenocarcinoma (n = 11) samples. Using absGSEA, we identified significant cellular pathways and putative molecular biomarkers associated with each stage of CRC progression. Key findings were then validated in a large independent CRC patient cohort (n = 1926), with survival analysis conducted from 1336 patients to assess the prognostic relevance of the candidate biomarkers. The key differentially expressed genes were experimentally validated using real-time PCR (RT-qPCR). Pathway analysis revealed that in CIS, apoptotic processes and Wnt signaling pathways were more prominent than in adenoma samples, while in adenocarcinoma, transcriptional co-regulatory mechanisms and protein kinase activity, which are critical for tumor growth and metastasis, were significantly enriched compared to adenoma. Additionally, extracellular matrix organization pathways were significantly enriched in adenocarcinoma compared to CIS. Distinct gene signatures were identified across CRC stages that differentiate between adenoma, CIS and adenocarcinoma. In adenoma, ARRB1, CTBP1 and CTBP2 were overexpressed, suggesting their involvement in early tumorigenesis, whereas in CIS, RPS3A and COL4A5 were overexpressed, suggesting their involvement in the transition from benign to malignant stage. In adenocarcinoma, COL1A2, CEBPZ, MED10 and PAWR were overexpressed, suggesting their involvement in advanced disease progression. Functional analysis confirmed that ARRB1 and CTBP1/2 were associated with early tumor development, while COL1A2 and CEBPZ were involved in extracellular matrix remodeling and transcriptional regulation, respectively. Experimental validation with RT-qPCR confirmed the differential expression of the candidate biomarkers (ARRB1, RPS3A, COL4A5, COL1A2 and MED10) across the three CRC stages reinforcing their potential as stage-specific biomarkers in CRC progression. These findings provide a foundation to distinguish between the CRC stages and for the development of accurate stage-specific diagnostic and prognostic biomarkers, which helps in the development of more effective therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Faisal Alhosani
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
- Forensic Laboratory Department, Sharjah Police Headquarters, Sharjah P.O. Box 1965, United Arab Emirates
| | - Reem Sami Alhamidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Burcu Yener Ilce
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Alaa Muayad Altaie
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Nival Ali
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
| | - Alaa Mohamed Hamad
- College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates;
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
| | - Cyrus Khandanpour
- Department of Hematology and Oncology, University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, University of Lübeck, 23562 Lübeck, Germany;
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; (A.K.); (H.B.)
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Rania Harati
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Kadria Sayed
- Department of Pathology and Laboratory Medicine, American Hospital Dubai, Dubai P.O. Box 3050, United Arab Emirates;
| | - Ali AlFazari
- Mediclinic Welcare Hospital, Dubai P.O. Box 31500, United Arab Emirates;
| | - Riyad Bendardaf
- Oncology Unit, University Hospital Sharjah, Sharjah P.O. Box 72772, United Arab Emirates;
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (F.A.); (R.S.A.); (B.Y.I.); (A.M.A.); (N.A.); (R.H.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
| |
Collapse
|
31
|
Liang Z, Li S, Wang Z, Zhou J, Huang Z, Li J, Bao H, Yam JWP, Xu Y. Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications. J Clin Transl Hepatol 2025; 13:315-326. [PMID: 40206274 PMCID: PMC11976435 DOI: 10.14218/jcth.2024.00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors in the world, and its incidence and mortality have increased year by year. HCC research has increasingly focused on understanding its pathogenesis and developing treatments.The Wnt signaling pathway, a complex and evolutionarily conserved signal transduction system, has been extensively studied in the genesis and treatment of several malignant tumors. Recent investigations suggest that the pathogenesis of HCC may be significantly influenced by dysregulated Wnt/β-catenin signaling. This article aimed to examine the pathway that controls Wnt signaling in HCC and its mechanisms. In addition, we highlighted the role of this pathway in HCC etiology and targeted treatment.
Collapse
Affiliation(s)
- Zixin Liang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shanshan Li
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyu Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junting Zhou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyue Huang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiehan Li
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haolin Bao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
32
|
Wu L, Li J, Wang H, Chang X, Kong Q. Identification of the oncogenic role of centromere protein M in non-small cell lung cancer via CDC20/MYBL2/Wnt signaling pathways. J Mol Histol 2025; 56:144. [PMID: 40285932 PMCID: PMC12033124 DOI: 10.1007/s10735-025-10423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/16/2024] [Indexed: 04/29/2025]
Abstract
Lung cancer remains the most prevalent carcinoma with a high mortality rate, yet the underlying mechanisms driving pulmonary neoplasia and disease progression are not fully understood. In our study, we conducted a comprehensive analysis of the transcriptome profiles and clinicopathological characteristics of 515 patients diagnosed with non-small cell lung cancer (NSCLC) from the TCGA database. We identified a significant upregulation of centromere protein M (CENPM) in NSCLC tissues, which was positively correlated with poor prognosis. Furthermore, overexpression of CENPM markedly promoted cell proliferation and increased the tumorigenic potential of NSCLC cell lines (A549/NCI-H1299), leading to accelerated tumor progression and reduced survival time in tumor-bearing mice. Mechanistically, CENPM activated the Wnt/β-catenin signaling pathway via the cell division cycle 20 (CDC20)/MYB proto-oncogene-like 2 (MYBL2) axis. Inhibition of either Wnt signaling or the CDC20/MYBL2 axis attenuated the tumorigenic potential and proliferative effects induced by CENPM. Our findings underscore the critical role of CENPM in driving NSCLC development and suggest that CENPM could serve as a novel biomarker for predicting NSCLC progression in clinical settings.
Collapse
Affiliation(s)
- Ling Wu
- Department of pharmacy, Affiliated Central Hospital of Dalian University of Technology, Dalian, China
| | - Jun Li
- Department of Thoracic Surgery, Affiliated Central Hospital of Dalian University of Technology, Dalian, China
| | - Haoyu Wang
- Department of Thoracic Surgery, Affiliated Central Hospital of Dalian University of Technology, Dalian, China
| | - Xu Chang
- Department of Thoracic Surgery, Affiliated Central Hospital of Dalian University of Technology, Dalian, China
| | - Qinglong Kong
- Department of Thoracic Surgery, Affiliated Central Hospital of Dalian University of Technology, Dalian, China.
| |
Collapse
|
33
|
Zhao K, Yan Y, Jin XK, Pan T, Zhang SM, Yang CH, Rao ZY, Zhang XZ. An orally administered gene editing nanoparticle boosts chemo-immunotherapy in colorectal cancer. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01904-5. [PMID: 40269250 DOI: 10.1038/s41565-025-01904-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 03/12/2025] [Indexed: 04/25/2025]
Abstract
Chemoresistance and immunosuppression are common obstacles to the efficacy of chemo-immunotherapy in colorectal cancer (CRC) and are regulated by mitochondrial chaperone proteins. Here we show that the disruption of the tumour necrosis factor receptor-associated protein 1 (TRAP1) gene, which encodes a mitochondrial chaperone in tumour cells, causes the translocation of cyclophilin D in tumour cells. This process results in the continuous opening of the mitochondrial permeability transition pore, which enhances chemotherapy-induced cell necrosis and promotes immune responses. On the basis of this discovery we developed an oral CRISPR-Cas9 delivery system based on zwitterionic and polysaccharide polymer-coated nanocomplexes that disrupts the TRAP1 gene in CRC. This system penetrates the intestinal mucus layer and undergoes epithelial transcytosis, accumulating in CRC tissues. It enhances chemotherapeutic efficacy by overcoming chemoresistance and activating the tumour immune microenvironment in orthotopic, chemoresistant and spontaneous CRC models, with remarkable synergistic antitumour effects. This oral CRISPR-Cas9 delivery system represents a promising therapeutic strategy for the clinical management of CRC.
Collapse
Affiliation(s)
- Kai Zhao
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Yu Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Xiao-Kang Jin
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Ting Pan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Shi-Man Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Chi-Hui Yang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Zhi-Yong Rao
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, P. R. China.
| |
Collapse
|
34
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
35
|
Luo Y, He Y, Xu Y, Wang Y, Yang L. The KDM5A/HOXA5 axis regulates osteosarcoma progression via activating the Wnt/β-catenin pathway. Eur J Med Res 2025; 30:284. [PMID: 40229896 PMCID: PMC11998425 DOI: 10.1186/s40001-025-02478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/19/2025] [Indexed: 04/16/2025] Open
Abstract
As an oncogenic driver, lysine-specific demethylase 5A (KDM5A) participates in regulating numerous tumor progression-related processes. Moreover, KDM5A functions as a histone demethylase, modulating the expression levels of its target genes by adjusting methylation levels. However, the underlying molecular mechanism of KDM5A in osteosarcoma remains elusive. To elucidate this mechanism, specifically how the KDM5A /Homeobox A5 (HOXA5) axis regulates osteosarcoma progression, we measured the expression levels of KDM5A and HOXA5 genes using reverse transcription-quantitative real-time PCR. The correlation between HOXA5 and KDM5A was analyzed via Pearson correlation analysis and further validated through chromatin immunoprecipitation-quantitative real-time PCR. Immunohistochemistry was conducted to determine the number of KDM5A-or HOXA5-positive cells present in osteosarcoma tissues. Additionally, Western blot analysis was utilized to quantify the protein levels of KDM5A, HOXA5, di- and tri-methylation of lysine 4 on histone H3, and β-catenin. Colony formation assays, wound healing assays and flow cytometry were used to detect cell proliferation, migration and apoptosis. The factors associated with the five-year survival rate of patients were analyzed. Our results illustrated that KDM5A was up-regulated in osteosarcoma and associated with a poor prognosis; KDM5A knockdown inhibited osteosarcoma cell proliferation and migration and promotes apoptosis. Subsequently, KDM5A knockdown induced HOXA5 expression by promoting di- and tri-methylation of lysine 4 on histone H3 demethylation, and HOXA5 overexpression inhibited osteosarcoma cell proliferation and migration, and promoted apoptosis by inhibiting the Wnt/β-catenin pathway. We finally proved that HOXA5 silence weakened the inhibitory effect of sh- KDM5A on osteosarcoma proliferation and migration and promoted apoptosis via activating Wnt/β-catenin pathway in vivo and in vitro. Our study demonstrated that the KDM5A /HOXA5 axis regulates osteosarcoma progression by activating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yi Luo
- Department of Spine Surgery, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, The No.161 of the Shaoshan South Road, Changsha City, Hunan Province, China.
| | - Youzhi He
- Department of Spine Surgery, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, The No.161 of the Shaoshan South Road, Changsha City, Hunan Province, China
| | - Yuxia Xu
- Department of Spine Surgery, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, The No.161 of the Shaoshan South Road, Changsha City, Hunan Province, China
| | - Yongfu Wang
- Department of Spine Surgery, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, The No.161 of the Shaoshan South Road, Changsha City, Hunan Province, China
| | - Li Yang
- Department of Spine Surgery, Hengyang Medical School, The Affiliated Changsha Central Hospital, University of South China, The No.161 of the Shaoshan South Road, Changsha City, Hunan Province, China
| |
Collapse
|
36
|
Rahmatallah Y, Glazko G. Improving data interpretability with new differential sample variance gene set tests. BMC Bioinformatics 2025; 26:103. [PMID: 40229677 PMCID: PMC11998189 DOI: 10.1186/s12859-025-06117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Gene set analysis methods have played a major role in generating biological interpretations of omics data such as gene expression datasets. However, most methods focus on detecting homogenous pattern changes in mean expression while methods detecting pattern changes in variance remain poorly explored. While a few studies attempted to use gene-level variance analysis, such approach remains under-utilized. When comparing two phenotypes, gene sets with distinct changes in subgroups under one phenotype are overlooked by available methods although they reflect meaningful biological differences between two phenotypes. Multivariate sample-level variance analysis methods are needed to detect such pattern changes. RESULTS We used ranking schemes based on minimum spanning tree to generalize the Cramer-Von Mises and Anderson-Darling univariate statistics into multivariate gene set analysis methods to detect differential sample variance or mean. We characterized the detection power and Type I error rate of these methods in addition to two methods developed earlier using simulation results with different parameters. We applied the developed methods to microarray gene expression dataset of prednisolone-resistant and prednisolone-sensitive children diagnosed with B-lineage acute lymphoblastic leukemia and bulk RNA-sequencing gene expression dataset of benign hyperplastic polyps and potentially malignant sessile serrated adenoma/polyps. One or both of the two compared phenotypes in each of these datasets have distinct molecular subtypes that contribute to within phenotype variability and to heterogeneous differences between two compared phenotypes. Our results show that methods designed to detect differential sample variance provide meaningful biological interpretations by detecting specific hallmark gene sets associated with the two compared phenotypes as documented in available literature. CONCLUSIONS The results of this study demonstrate the usefulness of methods designed to detect differential sample variance in providing biological interpretations when biologically relevant but heterogeneous changes between two phenotypes are prevalent in specific signaling pathways. Software implementation of the methods is available with detailed documentation from Bioconductor package GSAR. The available methods are applicable to gene expression datasets in a normalized matrix form and could be used with other omics datasets in a normalized matrix form with available collection of feature sets.
Collapse
Affiliation(s)
- Yasir Rahmatallah
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Galina Glazko
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| |
Collapse
|
37
|
Murtazina A, Jimenez-Martinez Y, Ruiz Alcala G, Marchal JA, Tarabayeva A, Bitanova E, Rakhimbayev I, McDougall GJ, Bishimbayeva N, Boulaiz H. In Vitro Inhibition of Colon Cancer Stem Cells by Natural Polysaccharides Obtained from Wheat Cell Culture. Polymers (Basel) 2025; 17:1048. [PMID: 40284312 PMCID: PMC12030112 DOI: 10.3390/polym17081048] [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: 02/13/2025] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Natural polysaccharides (PSs) have shown inhibitory effects on differentiated cancer cells (DCCs), but their activity against cancer stem cells (CSCs) remains poorly understood. Here, we report that PSs from wheat cell cultures (WCCPSs) inhibit the proliferation of both DCCs and CSCs derived from HCT-116 colorectal cancer cells. Among them, NA and DC fractions showed the strongest anti-CSC activity. NA, rich in xylose, was effective at lower concentrations, while DC, enriched in xylose and galacturonic acid (GalUA), exhibited higher potency, with a lower IC50 and preferential activity against CSCs at higher doses. WCCPSs reduced β-catenin levels, and some fractions also downregulated Ep-CAM, CD44, and c-Myc. Notably, DC increased caspase-3 without inducing cytochrome C and caspase-8 overexpression, suggesting a mechanism promoting CSC differentiation rather than apoptosis. Correlation analysis linked xylose content to reduced c-Myc expression, and GalUA levels to increased caspase-3. These results suggest that WCCPS bioactivity may be related to their monosaccharide composition. Overall, our findings support the potential of wheat-derived PSs as CSC-targeting agents that suppress self-renewal and promote differentiation, offering a promising approach to reduce tumor aggressiveness and recurrence.
Collapse
Affiliation(s)
- Alima Murtazina
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
| | - Yaiza Jimenez-Martinez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
| | - Gloria Ruiz Alcala
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
| | - Anel Tarabayeva
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
| | - Elmira Bitanova
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
| | | | - Gordon J. McDougall
- Plant Biochemistry and Food Quality Group, Environmental and Biochemical Sciences Department, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, UK;
| | - Nazira Bishimbayeva
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
- Research Institute for Problems of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Houria Boulaiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| |
Collapse
|
38
|
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
|
39
|
Duță C, Dogaru CB, Muscurel C, Stoian I. Nanozymes: Innovative Therapeutics in the Battle Against Neurodegenerative Diseases. Int J Mol Sci 2025; 26:3522. [PMID: 40332015 PMCID: PMC12026839 DOI: 10.3390/ijms26083522] [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: 03/01/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS) and Huntington's disease (HD), represent a significant challenge to global health due to their progressive nature and the absence of curative treatments. These disorders are characterized by oxidative stress, protein misfolding, and neuroinflammation, which collectively contribute to neuronal damage and death. Recent advancements in nanotechnology have introduced nanozymes-engineered nanomaterials that mimic enzyme-like activities-as promising therapeutic agents. This review explores the multifaceted roles of nanozymes in combating oxidative stress and inflammation in neurodegenerative conditions. By harnessing their potent antioxidant properties, nanozymes can effectively scavenge reactive oxygen species (ROS) and restore redox balance, thereby protecting neuronal function. Their ability to modify surface properties enhances targeted delivery and biocompatibility, making them suitable for various biomedical applications. In this review, we highlight recent findings on the design, functionality, and therapeutic potential of nanozymes, emphasizing their dual role in addressing oxidative stress and pathological features such as protein aggregation. This synthesis of current research underscores the innovative potential of nanozymes as a proactive therapeutic strategy to halt disease progression and improve patient outcomes in neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | - Corina Muscurel
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.D.); (C.B.D.); (I.S.)
| | | |
Collapse
|
40
|
Ma L, Li R, Li P, Yu W, Tang Z, Si L, Tian H. GINS1 facilitates the development of lung adenocarcinoma via Wnt/β-catenin activation. World J Surg Oncol 2025; 23:122. [PMID: 40197379 PMCID: PMC11974172 DOI: 10.1186/s12957-025-03786-2] [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: 11/24/2024] [Accepted: 03/29/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma(LUAD) is the primary reason for cancer-related deaths globally. GINS1 has a significant regulatory function in DNA replication. It is overexpressed in various malignant tumors, but the specific molecular mechanisms of GINS1 in LUAD pathogenesis are not fully elucidated. This is the first report that GINS1 enhances LUAD by activating Wnt/β-catenin signaling pathway, and may serve as a potential target for therapy. METHODS Bioinformatic analysis including analysis of difference, survival analysis and pathway enrichment, immunohistochemistry(IHC), western blotting(WB), and quantitative real time polymerase chain reaction(qRT-PCR) were used to detect GINS1 expression in LUAD cell lines and tissues. A range of in vivo and in vitro experiments, such as cck-8, EdU, cloning experiment, wound healing experiment and transwell experiment, confirmed that GINS1 facilitated the proliferation and migration of LUAD. Additionally, the potential mechanism of GINS1 was hypothesized through WB and transcriptome sequencing. The rescue experiment was used to verify our conclusion. RESULTS In this study, we discovered that GINS1 is significantly overexpressed in LUAD cell lines and tissues. Analysis of Kaplan - Meier survival data indicated that high levels of GINS1 expression are often linked to unfavorable survival outcomes. Additionally, a series of experiments showed that silencing GINS1 led to less proliferation and migration of LUAD cell lines, while its overexpression enhanced tumor progression. Furthermore, subcutaneous tumor experiments in nude mice supported the role of GINS1 in promoting tumor development in vivo. Lastly, transcriptome sequencing revealed that tumor progression is related to cell cycle (G1 to S phase transition associated with cyclinD) and β-catenin signaling pathway, which we subsequently validated using WB. A series of rescue experiment further confirmed that GINS1 facilitates the advancement of LUAD via the β-catenin signaling pathway. CONCLUSIONS Our findings suggest that GINS1 plays a critical role in the progression of LUAD by modulating key molecular pathways, particularly the β-catenin signaling pathway., and it might serve as a potential new target of β-catenin signaling pathway for treatment of LUAD.
Collapse
Affiliation(s)
- Luyuan Ma
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Rongyang Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Pengyong Li
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Wenhao Yu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhanpeng Tang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Libo Si
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
41
|
Samandar F, Mohsenpour A, Rastin F, Doustmohammadi-Salmani S, Saberi MR, Chamani J. Evaluating binding behavior of quercetin to human serum albumin and calf thymus DNA: Insights from molecular dynamics, spectroscopy, and apoptotic pathway regulation. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125638. [PMID: 39733709 DOI: 10.1016/j.saa.2024.125638] [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: 09/21/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
In this work, we sought to apprehend quercetin binding affinity and its interaction behavior in complex with human serum albumin (HSA) and calf thymus DNA (ctDNA) through multi spectroscopy and molecular dynamics and also evaluated its effects on colorectal cancer. The binding constants of ctDNA-quercetin and HSA-quercetin complexes at 298 K, which were calculated to be (2.67 ± 0.04) × 103 M-1 and (4.77 ± 0.05) × 104 M-1 respectively, denoted the strong binding of quercetin with ctDNA and HSA. The Ksv and Kb values decrease with increasing temperature, indicating that the quenching of HSA and ctDNA in the presence of quercetin is caused by the combined dynamic and static effects. The obtained thermodynamic parameters for the ctDNA-quercetin interaction represented the existence of electrostatic forces (ΔH0 < 0 and ΔS0 > 0), and the thermodynamic parameters of HSA-quercetin complex disclose the dominance of hydrogen bonds and van der Waals interactions (ΔH0 < 0 and ΔS0 < 0). Moreover, the interactions were exothermic, as evidenced by the negative ΔH0 value for both interactions. According to molecular docking and MD simulation data, quercetin was capable of placing into the site 1 of HSA and forming stable interaction plus this ligand tended to unwind DNA's strands as an intercalator ligand, which was confirmed by experimental results. The fluorescence competition studies between the two intercalator probes of ethidium bromide (EB) and acridine orange (AO), as well as the effect of ionic strength, proposed the strong tendency of quercetin to exist between the two strands of ctDNA as a sign of its intercalative property. Consequently, quercetin can be assumed as an efficient intercalator ligand carried by HSA with an anticancer property. We also conducted cell viability experiments on HT-29 and SW620 cell lines to validate the anticancer ability of quercetin, and observed its decreasing impact on the cell viability of these two cell lines. Additionally, the outcomes of Real-time qPCR proved its capability to reduce the CXCR4 expression and increase the NKD2 expression in Wnt signaling pathway. Therefore, these facts confirm the inhibiting ability of quercetin towards colorectal cancer growth via the prevention of Wnt pathway and approve its functionality as a potential anticancer agent for this cancer.
Collapse
Affiliation(s)
- Farzaneh Samandar
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Aida Mohsenpour
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Farangis Rastin
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Mohammad Reza Saberi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshidkhan Chamani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| |
Collapse
|
42
|
Li K, Dai YJ, Zhang H, Zhang Z. YAP1 activates SLC2A1 transcription and augments the malignant behavior of colorectal cancer cells by activating the Wnt/β-catenin signaling pathway. Cell Div 2025; 20:8. [PMID: 40186232 PMCID: PMC11969700 DOI: 10.1186/s13008-025-00148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This paper examined the role of solute carrier family 2 member 1 (SLC2A1) in colorectal cancer (CRC) progression, focusing on its expression levels, functional implications, and regulatory mechanisms involving Yes-associated protein 1 (YAP1) and the Wnt signaling pathway. METHODS GEO datasets (GSE14297, GSE18462, GSE40367) were analyzed to identify genes linked to metastasis in CRC, and TCGA-COAD system was used to analyze the expression pattern and prognostic values of SLC2A1 in CRC. Functional studies were conducted using CRC cell lines (Caco-2 and SW480). Cell viability, migration and invasion, and apoptosis were examined using EdU assays, Transwell assays, and flow cytometry. YAP1's regulatory role on SLC2A1 was investigated using ChIP-qPCR and luciferase reporter assays. The Wnt/β-catenin agonist SKL2001 was used for functional rescue experiments. RESULTS SLC2A1 was upregulated in CRC cells, and its upregulation was associated with tumor metastasis and unfavorable outcomes according to bioinformatics. Knockdown of SLC2A1 resulted in reduced cell viability, decreased migration, and increased apoptosis in Caco-2 and SW480 cells. Additionally, YAP1 was identified as a transcriptional activator of SLC2A1. Knockdown of YAP1 decreased SLC2A1 expression and reduced expression of Wnt target genes, thus suppressing malignant behavior of tumor cells. However, further overexpression of SLC2A1 restored cell viability and migration in YAP1-deficient cells. The YAP1- SLC2A1 axis activated the Wnt/β-catenin by reducing GSK3β activity. CONCLUSION SLC2A1 is critical in CRC progression, with YAP1 serving as a key regulator of its expression and function. The YAP1-SLC2A1-Wnt axis represents a potential therapeutic target for CRC, providing insights into metabolic adaptations that support tumor growth and metastasis.
Collapse
Affiliation(s)
- Kunpeng Li
- Zhongda Hospital of Southeast University, No 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, PR China
| | - Ya-Jie Dai
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Haifeng Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Zhigang Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China.
| |
Collapse
|
43
|
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
|
44
|
Tufail M, Jiang CH, Li N. Wnt signaling in cancer: from biomarkers to targeted therapies and clinical translation. Mol Cancer 2025; 24:107. [PMID: 40170063 PMCID: PMC11963613 DOI: 10.1186/s12943-025-02306-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: 02/04/2025] [Accepted: 03/15/2025] [Indexed: 04/03/2025] Open
Abstract
The Wnt signaling pathway plays a crucial role in development and tissue homeostasis, regulating key cellular processes such as proliferation, differentiation, and apoptosis. However, its abnormal activation is strongly associated with tumorigenesis, metastasis, and resistance to therapy, making it a vital target for cancer treatment. This review provides a comprehensive insight into the role of Wnt signaling in cancer, examining its normal physiological functions, dysregulation in malignancies, and therapeutic potential. We emphasize the importance of predicting Wnt signaling sensitivity and identify key biomarkers across various cancer types. Additionally, we address the challenges and future prospects of Wnt-targeted therapies, including biomarker discovery, advancements in emerging technologies, and their application in clinical practice.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
45
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
46
|
Zhang QQ, Miao YS, Hu JY, Liu RX, Hu YX, Wang F. The truncated AXIN1 isoform promotes hepatocellular carcinoma metastasis through SRSF9-mediated exon 9 skipping. Mol Cell Biochem 2025; 480:2247-2263. [PMID: 38748384 DOI: 10.1007/s11010-024-05012-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/14/2024] [Indexed: 04/02/2025]
Abstract
Axis inhibitor protein 1 (AXIN1) is a protein recognized for inhibiting tumor growth and is commonly involved in cancer development. In this study, we explored the potential molecular mechanisms that connect alternative splicing of AXIN1 to the metastasis of hepatocellular carcinoma (HCC). Transcriptome sequencing, RT‒PCR, qPCR and Western blotting were utilized to determine the expression levels of AXIN1 in human HCC tissues and HCC cells. The effects of the AXIN1 exon 9 alternative splice isoform and SRSF9 on the migration and invasion of HCC cells were assessed through wound healing and Transwell assays, respectively. The interaction between SRSF9 and AXIN1 was investigated using UV crosslink RNA immunoprecipitation, RNA pulldown, and RNA immunoprecipitation assays. Furthermore, the involvement of the AXIN1 isoform and SRSF9 in HCC metastasis was validated in a nude mouse model. AXIN1-L (exon 9 including) expression was downregulated, while AXIN1-S (exon 9 skipping) was upregulated in HCC. SRSF9 promotes the production of AXIN1-S by interacting with the sequence of exons 8 and 10 of AXIN1. AXIN1-S significantly promoted HCC cells migration and invasion by activating the Wnt pathway, while the opposite effects were observed for AXIN1-L. In vivo experiments demonstrated that AXIN1-L inhibited HCC metastasis, whereas SRSF9 promoted HCC metastasis in part by regulating the level of AXIN1-S. AXIN1, a tumor suppressor protein that targets the AXIN1/Wnt/β-catenin signaling axis, may be a promising prognostic factor and a valuable therapeutic target for HCC.
Collapse
Affiliation(s)
- Qian-Qian Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Genomic Medicine, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Ying-Shuang Miao
- International School, Jinan University, Guangzhou, 510632, China
| | - Jun-Yi Hu
- International School, Jinan University, Guangzhou, 510632, China
| | - Rui-Xuan Liu
- International School, Jinan University, Guangzhou, 510632, China
| | - Yue-Xiao Hu
- International School, Jinan University, Guangzhou, 510632, China
| | - Feng Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Genomic Medicine, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
47
|
Mahajan M, Dhabalia S, Dash T, Sarkar A, Mondal S. A comprehensive multi-omics study reveals potential prognostic and diagnostic biomarkers for colorectal cancer. Int J Biol Macromol 2025; 303:140443. [PMID: 39909246 DOI: 10.1016/j.ijbiomac.2025.140443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/11/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND AND OBJECTIVE Colorectal cancer (CRC) is a complex disease with diverse genetic alterations and causes 10 % of cancer-related deaths worldwide. Understanding its molecular mechanisms is essential for identifying potential biomarkers and therapeutic targets for its effective management. METHODS We integrated copy number alterations (CNA) and mutation data via their differentially expressed genes termed as candidate genes (CGs) computed using bioinformatics approaches. Then, using the CGs, we perform Weighted correlation network analysis (WGCNA) and utilise several hazard models such as Univariate Cox, Least Absolute Shrinkage and Selection Operator (LASSO) Cox and multivariate Cox to identify the key genes involved in CRC progression. We used different machine-learning models to demonstrate the discriminative power of selected hub genes among normal and CRC (early and late-stage) samples. RESULTS The integration of CNA with mRNA expression identified over 3000 CGs, including CRC-specific driver genes like MYC and APC. In addition, pathway analysis revealed that the CGs are mainly enriched in endocytosis, cell cycle, wnt signalling and mTOR signalling pathways. Hazard models identified four key genes, CASP2, HCN4, LRRC69 and SRD5A1, that were significantly associated with CRC progression and predicted the 1-year, 3-years, and 5-years survival times. WGCNA identified seven hub genes: DSCC1, ETV4, KIAA1549, NOP56, RRS1, TEAD4 and ANKRD13B, which exhibited strong predictive performance in distinguishing normal from CRC (early and late-stage) samples. CONCLUSIONS Integrating regulatory information with gene expression improved early versus late-stage prediction. The identified potential prognostic and diagnostic biomarkers in this study may guide us in developing effective therapeutic strategies for CRC management.
Collapse
Affiliation(s)
- Mohita Mahajan
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K.K. Birla Goa campus, Zuarinagar, Goa 403726, India.
| | - Subodh Dhabalia
- Department of Mathematics, Amrita Vishwa Vidyapeetham, Amritanagar, Coimbatore 64112, India.
| | - Tirtharaj Dash
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
| | - Angshuman Sarkar
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K.K. Birla Goa campus, Zuarinagar, Goa 403726, India.
| | - Sukanta Mondal
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K.K. Birla Goa campus, Zuarinagar, Goa 403726, India.
| |
Collapse
|
48
|
Yuan J, Zhang K, Yang L, Cheng X, Chen J, Guo X, Cao H, Zhang C, Xing C, Hu G, Zhuang Y. Luteolin attenuates LPS-induced damage in IPEC-J2 cells by enhancing mitophagy via AMPK signaling pathway activation. Front Nutr 2025; 12:1552890. [PMID: 40206944 PMCID: PMC11978636 DOI: 10.3389/fnut.2025.1552890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
Background Luteolin (LUT), a flavonoid compound widely present in natural plants, has been extensively studied for its diverse biological properties, involving anti-inflammatory,antioxidant, anti-apoptosis and other properties. Methods The aim of this study was to investigate the effect of LUT on lipopolysaccharide (LPS)-induced Intestinal Porcine Epithelial Cell line-J2 (IPEC-J2 cells) damage and its underlying mechanism. Results The experiment showed that LPS treatment induced injury in IPEC-J2 cells, leading to tight junction disruption, ROS accumulation, and cell apoptosis. Remarkably, LUT attenuated LPS-induced IPEC-J2 cells damage by the up-regulation of Zonula Occludens-1(ZO-1), Occludin, and Claudin protein 1 (Claudin-1) protein expression levels.Besides, LUT increased the activities of CAT, and SOD and prevented LPS-induced MDA and ROS production. LUT suppressed Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation in LPS-induced IPEC-J2 cells, reducing (Interleukin-1beta) IL-1β and Interleukin-6 (IL-6) expression. Moreover, LUT attenuated LPS-induced apoptosis in IPEC-J2 cells by up-regulating expression of B-cell lymphoma 2 (Bcl-2) and down-regulating expression of Cysteine-aspartic acid protease 3 (Caspase-3), Cysteine - aspartic acid protease 9 (Caspase-9) and Bcl-2-associated X protein (Bax). Furthermore, LUT upregulated the AMP-activated protein kinase (AMPK)/Unc-51 like autophagy activating kinase (ULK) signaling pathway and Parkin-RBR E3 ubiquitin protein ligase (Parkin)/PTEN induced putative kinase 1 (PINK1)-mediated mitophagy in a dose-dependent manner. When AMPK was knocked down by short-hairpin RNA (shRNA), the protective effects of LUT against LPS-induced IPEC-J2 cell damage were weakened, as evidenced by the accumulation of excessive ROS and impaired mitophagy. Conclusion In summary, LUT exhibits the ability to protect against LPS-induced damage to intestinal tight junctions by enhancing mitophagy through AMPK activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| |
Collapse
|
49
|
Chen Q, Wang H, Liu Q, Luo C. CTHRC1: a key player in colorectal cancer progression and immune evasion. Front Immunol 2025; 16:1579661. [PMID: 40201173 PMCID: PMC11975584 DOI: 10.3389/fimmu.2025.1579661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
The multifunctional secreted protein, collagen triple helix repeat containing 1 (CTHRC1), has recently emerged as a significant focus within oncology research. CTHRC1 expression in tumors is governed by a complex interplay of regulatory signals, including methylation, glycosylation, and notably, non-coding RNAs, which constitute its predominant regulatory mechanism. Colorectal cancer (CRC), a highly prevalent epithelial malignancy, sees CTHRC1 influencing tumor progression and metastasis through its modulation of several downstream signaling cascades, such as Wnt/PCP, TGF-β/Smad, and MEK/ERK pathways. Furthermore, CTHRC1 contributes to immune evasion in CRC via diverse mechanisms. It is intricately associated with macrophage phenotypic switching within the tumor microenvironment (TME), favoring M2 macrophage polarization and facilitating the infiltration of T cells and neutrophils. CTHRC1 is also instrumental in immune escape by driving the remodeling of the extracellular matrix through interactions with cancer-associated fibroblasts. Additionally, CTHRC1's roles extend to the regulation of hypoxia-related pathways, metabolism of glycolysis and fatty acids, and involvement in tumor angiogenesis, all of which support tumor immune evasion. Considering its multifaceted activities, CTHRC1 emerges as a promising therapeutic target in CRC, with the potential to enhance the outcomes of existing radiotherapeutic and immunotherapeutic regimens. This review endeavors to delineate the mechanistic and therapeutic landscapes of CTHRC1 in CRC. Through a comprehensive discussion of CTHRC1's diverse functions, we aim to provide insights that could pave the way for innovative approaches in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Changjiang Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
50
|
Obisi JN, Abimbola ANJ, Babaleye OA, Atidoglo PK, Usin SG, Nwanaforo EO, Patrick-Inezi FS, Fasogbon IV, Chimezie J, Dare CA, Kuti OO, Uti DE, Omeoga HC. Unveiling the future of cancer stem cell therapy: a narrative exploration of emerging innovations. Discov Oncol 2025; 16:373. [PMID: 40120008 PMCID: PMC11929669 DOI: 10.1007/s12672-025-02102-4] [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] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer stem cells (CSCs), are a critical subpopulation within tumours, and are defined by their capacity for self-renewal, differentiation, and tumour initiation. These unique traits contribute to tumour progression, metastasis, and resistance to conventional treatments like chemotherapy and radiotherapy, often resulting in cancer recurrence and poor patient outcomes. As such, CSCs have become focal points in developing advanced cancer therapies. This review highlights progress in CSC-targeted treatments, including chimeric antigen receptor T-cell (CAR-T) therapy, immunotherapy, molecular targeting, and nanoparticle-based drug delivery systems. Plant-derived compounds and gene-editing technologies, such as clustered regularly interspaced short palindromic repeats (CRISPR), are explored for their potential to enhance precision and minimize side effects. Metabolic pathways integral to CSC survival, such as mitochondrial dynamics, mitophagy (regulated by dynamin-related protein 1 [DRP1] and the PINK1/Parkin pathway), one-carbon metabolism, amino acid metabolism (involving enzymes like glutaminase (GLS) and glutamate dehydrogenase (GDH]), lipid metabolism, and hypoxia-induced metabolic reprogramming mediated by hypoxia-inducible factors (HIF-1α and HIF-2α), are examined as therapeutic targets. The adaptability of CSCs through autophagy, metabolic flexibility, and epigenetic regulation by metabolites like α-ketoglutarate, succinate, and fumarate is discussed. Additionally, extracellular vesicles and nicotinamide adenine dinucleotide (NAD⁺) metabolism are identified as pivotal in redox balance, DNA repair, and epigenetic modifications. Addressing challenges such as tumour heterogeneity, immune evasion, and treatment durability requires interdisciplinary collaboration. Advancing CSC-targeted therapies is essential for overcoming drug resistance and preventing cancer relapse, paving the way for transformative cancer treatments. This review underscores the importance of leveraging innovative technologies and fostering collaboration to revolutionize cancer treatment.
Collapse
Affiliation(s)
| | | | - Oluwasegun Adesina Babaleye
- Center for Human Virology and Genomics, Department of Microbiology, Nigerian Institute of Medical Research, Lagos, Nigeria
| | - Peter Kwame Atidoglo
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Saviour God'swealth Usin
- Cancer Research and Molecular Biology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Eudora Obioma Nwanaforo
- Environmental Health Science Department, School of Heath Technology, Federal University of Technology Owerri, Owerri, Nigeria
| | | | | | - Joseph Chimezie
- Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | | | - Daniel Ejim Uti
- Department of Biochemistry/Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Benue State, Nigeria.
| | | |
Collapse
|