1
|
Xiong X, Du Y, Liu P, Li X, Lai X, Miao H, Ning B. Unveiling EIF5A2: A multifaceted player in cellular regulation, tumorigenesis and drug resistance. Eur J Pharmacol 2025; 997:177596. [PMID: 40194645 DOI: 10.1016/j.ejphar.2025.177596] [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: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
The eukaryotic initiation factor 5A2 gene (EIF5A2) is a highly conserved and multifunctional gene that significantly influences various cellular processes, including translation elongation, RNA binding, ribosome binding, protein binding and post-translational modifications. Overexpression of EIF5A2 is frequently observed in multiple cancers, where it functions as an oncoprotein. Additionally, EIF5A2 is implicated in drug resistance through the regulation of various molecular pathways. In the review, we describe the structure and functions of EIF5A2 in normal cells and its role in tumorigenesis. We also elucidate the molecular mechanisms associated with EIF5A2 in the context of tumorigenesis and drug resistance. We propose that the biological roles of EIF5A2 in regulating diverse cellular processes and tumorigenesis are clinically significant and warrant further investigation.
Collapse
Affiliation(s)
- Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China; Guangzhou Institute of Burn Clinical Medicine, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Yanli Du
- Guangdong Medical University, Zhanjiang, 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Peng Liu
- Departments of Burn and Plastic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Xinye Li
- Guangdong Medical University, Zhanjiang, 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Xudong Lai
- Department of infectious disease, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China
| | - Haixiong Miao
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China.
| | - Bo Ning
- Department of Neurosurgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, Guangdong, China.
| |
Collapse
|
2
|
Javaid D, Ganie SY, Qadri SS, Reyaz A, Reshi MS. Eco-friendly nanotherapeutics: Metallic nanoparticles for targeting breast cancer. Eur J Pharmacol 2025; 996:177603. [PMID: 40189083 DOI: 10.1016/j.ejphar.2025.177603] [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: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/13/2025]
Abstract
Breast cancer continues to be a major cause of death among women globally, with triple-negative breast cancer (TNBC) presenting a particularly difficult challenge due to its aggressive behaviour and the lack of effective treatment options. Nanotechnology, particularly the use of silver nanoparticles (AgNPs), has emerged as a promising avenue in oncological research. This review explores into the escalating field of green synthesis of nanoparticles, emphasizing sustainable approaches utilizing plant-based resources. Critical factors influencing nanoparticle synthesis, including reaction conditions, precursor types, and plant phytochemicals, are explored alongside advanced characterization techniques essential for evaluating nanoparticle properties. Special focus is given to the phytofabrication of silver nanoparticles and their multifaceted roles in breast cancer treatment, with detailed insights into their mechanisms, such as inducing apoptosis, generating reactive oxygen species (ROS), and disrupting mitochondrial function, particularly in TNBC cells. The review further highlights the advantages of plant-derived AgNPs, such as biocompatibility and reduced toxicity, while addressing challenges like scalability, reproducibility, and regulatory hurdles. Concluding with future prospects, this paper reflects the potential of green-synthesized AgNPs as a keystone in next-generation cancer therapeutics, paving the way for innovative and eco-friendly approaches in oncology.
Collapse
Affiliation(s)
- Darakhshan Javaid
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Shahid Yousuf Ganie
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Syed Sanober Qadri
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Adfar Reyaz
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Mohd Salim Reshi
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India.
| |
Collapse
|
3
|
Hossain MT, Hossain MA. Targeting PI3K in cancer treatment: A comprehensive review with insights from clinical outcomes. Eur J Pharmacol 2025; 996:177432. [PMID: 40020984 DOI: 10.1016/j.ejphar.2025.177432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway plays a crucial role in cancer, including cell growth, survival, metabolism, and metastasis. Its major role in tumor growth makes it a key target for cancer therapeutics, offering significant potential to slow tumor progression and enhance patient outcomes. Gain-of-function mutations, gene amplifications, and the loss of regulatory proteins like PTEN are frequently observed in malignancies, contributing to tumor development and resistance to conventional treatments such as chemotherapy and hormone therapy. As a result, PI3K inhibitors have received a lot of interest in cancer research. Several kinds of small-molecule PI3K inhibitors have been developed, including pan-PI3K inhibitors, isoform-specific inhibitors, and dual PI3K/mTOR inhibitors, each targeting a distinct component of the pathway. Some PI3K inhibitors such as idelalisib, copanlisib, duvelisib, alpelisib, and umbralisib have received FDA-approval, and are effective in the treatment of breast cancer and hematologic malignancies. Despite promising results in preclinical and clinical trials, the overall clinical success of PI3K inhibitors has been mixed. While some patients may get substantial advantages, a considerable number of them acquire resistance as a result of feedback activation of alternative pathways, adaptive tumor responses, and treatment-emergent mutations. The resistance mechanisms provide barriers to the sustained efficacy of PI3K-targeted treatments. This study reviews recent advancements in PI3K inhibitors, covering their clinical status, mechanism of action, resistance mechanisms, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Md Takdir Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| | - Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
4
|
Luo W, Sun Y, Cao L. TSPAN31 Activates Fatty Acid Metabolism and PI3K/AKT Pathway to Promote Tumor Progression in Breast Cancer. Mol Carcinog 2025; 64:1078-1089. [PMID: 40135650 DOI: 10.1002/mc.23912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
Breast cancer (BC) is one of the most common human malignancies, but the mechanisms of BC have not been fully elucidated. Recently, tetraspanin 31 (TSPAN31) is reported to be linked to cancer progression. However, the function of TSPAN31 remains unclear in BC. Investigation of the function and potential mechanism of TSPAN31 in BC was the purpose of this study. Immunohistochemistry, western blot, and quantitative real-time polymerase chain reaction were applied to measure TSPAN31 expression. Loss and gain functional experiments were utilized to survey the influences of TSPAN31 on BC biological process, including cell growth, invasion, migration, and fatty acid metabolism. Mechanistically, Kyoto Encyclopedia of Genes and Genomes analysis based on DepMap database and Gene Set Enrichment Analysis based on The Cancer Genome Atlas database were executed to find TSPAN31-related pathway. Western blot was carried out to assess the changes of fatty acid synthase (FASN), sterol regulatory element binding protein 1 (SREBP1), acyl-CoA synthetase long-chain family member 1 (ACSL1), phosphatidylinositol 3-kinase (PI3K), phosphorylated (p)-PI3K, protein kinase B (AKT), and p-AKT. In human non-triple negative breast cancer tissues and cells, TSPAN31 expression was upregulated. TSPAN31 knockdown induced BC cell apoptosis, inhibited cell proliferation, invasion, migration, and fatty acid metabolism, and reduced the protein levels of FASN, SREBP1, ACSL1, p-PI3K/PI3K, and p-AKT/AKT. In contrast, TSPAN31 overexpression led to the opposite results. Additionally, the activator of PI3K (740 Y-P) attenuated the inhibition of TSPAN31 knockdown on fatty acid metabolism, proliferation, and invasion in BC cells. Through activation of fatty acid metabolism and PI3K/AKT pathway, TSPAN31 played a carcinogenic role in BC. For the mechanism of BC tumorigenesis, our study provides an interesting insight.
Collapse
Affiliation(s)
- Wenquan Luo
- Breast and Thyroid Surgery Department, Feicheng People's Hospital, Feicheng, Shandong, China
| | - Yuxiang Sun
- Breast and Thyroid Surgery Department, Feicheng People's Hospital, Feicheng, Shandong, China
| | - Liang Cao
- Radiotherapy Department, Taian Tumor Prevention and Treatment Hospital, Taian, Shandong, China
| |
Collapse
|
5
|
Expert Consensus on the Clinical Application of PI3K/AKT/mTOR Inhibitors in the Treatment of Breast Cancer (2025 Edition). CANCER INNOVATION 2025; 4:e70008. [PMID: 40206206 PMCID: PMC11981814 DOI: 10.1002/cai2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Background The phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB or AKT)/mammalian target of rapamycin (mTOR) signaling pathway (PAM pathway) plays a critical role in breast cancer pathogenesis and progression, and is closely linked with resistance to endocrine therapy in advanced breast cancer. Randomized clinical trials have shown that PI3K/AKT/mTOR inhibitors deliver significant clinical benefits, particularly for patients with advanced hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer. Methods In 2022, the Breast Cancer Expert Committee of the National Cancer Quality Control Center convened specialists in related fields to draft the "Expert Consensus on the Clinical Application of PI3K/AKT/mTOR Inhibitors in the Treatment of Advanced Breast Cancer." This consensus raised awareness of these inhibitors among oncologists in China and improved the precision of clinical decision-making. In recent years, growing evidence has emphasized the importance of targeting the PAM pathway, reflected in the approval of several innovative agents. This consensus is an updated 2025 edition that retains the foundational structure of the 2022 edition while incorporating notable updates. Results Updates to the consensus include the introduction of newly approved PAM pathway inhibitors, updated data from recent clinical trials, and expanded therapeutic applications. The revised guidance also offers updated recommendations for genetic testing to detect alterations in relevant pathways. The section on managing drug-related adverse events has been significantly expanded, providing detailed insights into different types of adverse events and their management. These updates aim to enhance the clinical application of PAM pathway inhibitors, promote precision medicine, and ultimately, improve survival outcomes for patients with breast cancer.
Collapse
|
6
|
Garg P, Ramisetty S, Nair M, Kulkarni P, Horne D, Salgia R, Singhal SS. Strategic advancements in targeting the PI3K/AKT/mTOR pathway for Breast cancer therapy. Biochem Pharmacol 2025; 236:116850. [PMID: 40049296 DOI: 10.1016/j.bcp.2025.116850] [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: 12/11/2024] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Breast cancer (BC) is a complex disease that affects millions of women worldwide. Its growing impact calls for advanced treatment strategies to improve patient outcomes. The PI3K/AKT/mTOR pathway is a key focus in BC therapy because it plays a major role in important processes like tumor growth, survival, and resistance to treatment. Targeting this pathway could lead to better treatment options and outcomes. The present review explores how the PI3K/AKT/mTOR pathway becomes dysregulated in BC, focusing on the genetic changes like PIK3CA mutations and PTEN loss that leads to its aggravation. Current treatment options include the use of inhibitors targeting PI3K, AKT, and mTOR with combination therapies showing promise in overcoming drug resistance and improving effectiveness. Looking ahead, next-generation inhibitors and personalized treatment plans guided by biomarker analysis may provide more accurate and effective options for patients. Integrating these pathway inhibitors with immunotherapy offers an exciting opportunity to boost anti-tumor responses and improve survival rates. This review offers a comprehensive summary of the current progress in targeting the PI3K/AKT/mTOR pathway in BC. It highlights future research directions and therapeutic strategies aimed at enhancing patient outcomes and quality of life.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sravani Ramisetty
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Meera Nair
- William J. Brennan High School, San Antonio, TX 78253, USA
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S Singhal
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
7
|
Chen Y, Bao S, Ding Y, Weng G, Zheng S, Ge C, Zhang C. Colchicine inhibits myocardial pyroptosis and reduces myocardial cell injury after myocardial infarction through the ESR1-PI3K-Akt-NF-κB signaling pathway. Int Immunopharmacol 2025; 156:114732. [PMID: 40294468 DOI: 10.1016/j.intimp.2025.114732] [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: 12/09/2024] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND AND PURPOSE Inflammation serves as a critical driver in coronary artery disease pathogenesis. Emerging clinical evidence demonstrates that low-dose colchicine therapy significantly reduces ischemic event incidence in patients with coronary heart disease while attenuating myocardial ischemia-induced inflammatory cascades. Nevertheless, the precise cardioprotective mechanisms underlying colchicine-a plant-derived anti-inflammatory agent-in limiting post-infarction cardiomyocyte injury remain incompletely elucidated. This study systematically investigates colchicine's myocardial preservation mechanisms through an integrated experimental approach. METHOD To establish experimental models of myocardial injury, we performed permanent ligation of the left anterior descending coronary artery (LAD) in mice for in vivo studies, while HL-1 mouse atrial cardiomyocytes were treated with 0.3 mM H₂O₂ to induce oxidative stress in vitro. Following successful model validation, colchicine was administered to both systems. Comprehensive evaluations included echocardiographic assessment of cardiac function, histological examination of inflammatory infiltration and collagen deposition through H&E and Masson's trichrome staining respectively, quantitative analysis of cardiomyocyte apoptosis by flow cytometry, and Western blot detection of key signaling pathway components and pyroptosis-related proteins (including NLRP3, caspase-1, and GSDMD). RESULT Our experimental data revealed that colchicine treatment significantly attenuated myocardial injury and fibrosis while improving cardiac function (P < 0.05). Mechanistically, colchicine administration reduced proinflammatory cytokine release (IL-1β and IL-18), decreased neutrophil infiltration, and suppressed cardiomyocyte pyroptosis. These cardioprotective effects were associated with modulation of the ESR1-PI3K-Akt-NF-κB signaling pathway (P < 0.05), suggesting a potential therapeutic mechanism for colchicine in myocardial protection. CONCLUSION Colchicine inhibits myocardial pyroptosis and reduces myocardial cell injury after myocardial infarction through the ESR1-PI3K-Akt-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yao Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - ShanQing Bao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yijie Ding
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
| | - GuangDong Weng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - ShiJie Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - ChaoLiang Ge
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, China
| | - ChengXin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| |
Collapse
|
8
|
Han R, Gao C, Tang R, Gui X, Chen W, Fu J, Wang T, Bai D, Guo Y, Zhou C. A comprehensive study on Herba Epimedium-derived extracellular nanovesicles as a prospective therapy for alveolar bone regeneration in postmenopausal osteoporosis. NANOSCALE 2025; 17:12270-12289. [PMID: 40266676 DOI: 10.1039/d5nr00508f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Natural products rich in phytoestrogens, particularly those derived from traditional Chinese medicine (TCM) herbs, have garnered increased attention. Plant-derived extracellular vesicles are emerging as a promising strategy in cell communication and disease defense. Here, a comprehensive study on Herba Epimedium-derived extracellular nanovesicles (EELNs) for postmenopausal osteoporosis treatment was conducted. The results showed that EELNs exhibit a typical exosome morphology with an average diameter of 130 nm and are rich in specific small-molecule metabolites and miRNAs. Network pharmacology and KEGG analysis highlighted the therapeutic potential of EELNs in osteoporosis through multiple classical osteogenic pathways. In vitro experiments proved that EELNs potentiated the osteogenic differentiation of BMSCs by targeting the Pi3k/Akt/mTOR pathway. In vivo, EELN-loaded hydroxyapatite nano-whisker (E-GW) composites were used to repair mandibular defects in an OVX-induced osteoporosis rat model. The results indicate that EELNs are promising therapeutic agents for the regeneration and bone mass maintenance of alveolar defects in postmenopausal osteoporosis patients and offer potential perspectives for natural products in postmenopausal osteoporosis treatment.
Collapse
Affiliation(s)
- Ruiying Han
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Canyu Gao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China.
| | - Rong Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Xingyu Gui
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China.
| | - Wanxi Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jiarun Fu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Tianyi Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Ding Bai
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yongwen Guo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
9
|
Ding T, Zhao S, Gu Y, He G, Lang Y, Rao X, Chen J, Ou-Yang Y. IL-17A regulates airway remodelling in COPD through the PI3K/AKT/mTOR pathway. Sci Rep 2025; 15:16546. [PMID: 40360557 PMCID: PMC12075786 DOI: 10.1038/s41598-025-00458-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The airway and lung tissue inflammation and structural changes caused by COPD lead to persistent and irreversible airflow limitation in patients. Several studies have associated IL-17A with COPD airway inflammation and collagen deposition, while autophagy is essential for maintaining normal cell function. Based on these findings, we propose a hypothesis that IL-17 affecting the autophagy of macrophages through the PI3K/AKT/mTOR pathway may contribute to the regulation of the airway remodeling process in COPD. The COPD airway remodelling model was confirmed by pulmonary function tests and HE and Masson staining. IL-17A, IL-6 and CCL20 were detected by ELISA, autophagosomes (APs) were observed by transmission electron microscopy, and western blotting was used to detect PI3K/AKT/mTOR-related proteins, autophagy-related proteins and collagen. Immunofluorescence revealed colocalization of LC3 in mouse bronchial fibroblasts (MBFs). MBFs were isolated and cultured via lentiviral transfection, IL-17RA overexpression or silencing, and quantitative analysis of PI3K/AKT/mTOR pathway-related proteins and autophagy-related proteins via western blotting. The results validated the establishment of the COPD model. Increased IL-17A in the COPD airway and increased IL-6 and CCL20 expression were observed in the COPD mice supplemented with the autophagy inhibitor 3MA. Using TEM, we observed a significant reduction in the number of APs in the airways in both the COPD and 3MA groups. Moreover, the PI3K/AKT/mTOR pathway phospho-p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR ratios were also increased in the COPD group. Moreover, as P62 increased, Beclin-1 and LC3II/I expression decreased correspondingly, while Collagen I and Collagen III also increased. In our study of cultured mouse MBFs, the results showed that overexpression (OE) virus-mediated transfection of MBFs overexpressing IL-17RA increased the p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR ratios, increased the P62 content and reduced the expression of Beclin-1 and LC3II/I. However, compared with the OE group, the silencing (sh)-mediated transfection of MBFs with IL-17RA had almost opposite effects. Increased collagen production and the expression of IL-17A, IL-6 and CCL20 in the airways of COPD mice. Autophagy decreased, and the PI3K/AKT/mTOR pathway was activated in COPD airway tissue. In primary cultured MBFs from COPD mice, IL-17A combined with IL-17RA activates the PI3K/AKT/mTOR pathway, thereby inhibiting autophagy.
Collapse
Affiliation(s)
- Ting Ding
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China.
| | - Shunshun Zhao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China
| | - Yanhui Gu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China
| | - Guiqiang He
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China
| | - Yanzhu Lang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China
| | - Ximin Rao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China
| | - Jie Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China
| | - Yao Ou-Yang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Guizhou, 563003, China.
| |
Collapse
|
10
|
Emami A, Mahdavi Sharif P, Rezaei N. KRAS mutations in colorectal cancer: impacts on tumor microenvironment and therapeutic implications. Expert Opin Ther Targets 2025:1-23. [PMID: 40320681 DOI: 10.1080/14728222.2025.2500426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION Despite decreasing trends in incidence, colorectal cancer (CRC) is still a major contributor to malignancy-related morbidities and mortalities. Groundbreaking advances in immunotherapies and targeted therapies benefit a subset of CRC patients, with sub-optimal outcomes. Hence, there is an unmet need to design and manufacture novel therapies, especially for advanced/metastatic disease. KRAS, the most highly mutated proto-oncogene across human malignancies, particularly in pancreatic adenocarcinoma, non-small cell lung cancer, and CRC, is an on-off switch and governs several fundamental cell signaling cascades. KRAS mutations not only propel the progression and metastasis of CRC but also critically modulate responses to targeted therapies. AREAS COVERED We discuss the impacts of KRAS mutations on the CRC's tumor microenvironment and describe novel strategies for targeting KRAS and its associated signaling cascades and mechanisms of drug resistance. EXPERT OPINION Drug development against KRAS mutations has been challenging, mainly due to structural properties (offering no appropriate binding site for small molecules), critical functions of the wild-type KRAS in non-cancerous cells, and the complex network of its downstream effector pathways (allowing malignant cells to develop resistance). Pre-clinical and early clinical data offer promises for combining KRAS inhibitors with immunotherapies and targeted therapies.
Collapse
Affiliation(s)
- Anita Emami
- Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nima Rezaei
- Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Prajoko YW, Heriyanto DS, Dirja BT, Susanto S, Lau V, Gunawan AN, Halim BN, Amalina ND. High frequency of the PIK3CA H1047L mutation in Indonesian breast cancer across molecular subtypes. PLoS One 2025; 20:e0322154. [PMID: 40323930 PMCID: PMC12052133 DOI: 10.1371/journal.pone.0322154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025] Open
Abstract
Breast cancer (BC) is a global health concern with significant mortality rates, necessitating a deep understanding of its molecular landscape. Luminal A and B BC, characterized by estrogen receptor (ER) and/or progesterone receptor (PR) positivity, face challenges in endocrine therapy due to acquired resistance, frequently driven by PI3K/AKT/mTOR pathway activation. This study focuses on the frequency of PIK3CA mutations across molecular subtypes BC within the Indonesian population. The study analyzed collected samples from diverse Indonesian regions, representing various islands. Histopathological analysis and immunohistochemistry classified samples into molecular subtypes. Genetic analysis using PIK3CA mutation detection kits revealed a mutation frequency of 32.9%, with 30 (14.5%) samples located in exon 9 and 38 (18.4%) samples in exon 20. Statistical analyses highlighted associations between PIK3CA mutations and molecular subtypes (p = 0.029), with luminal B HER2-negative (40.5%) and luminal A (40.2%) exhibiting the highest mutation rate. A significant association was also observed between the exon location of only mutated PIK3CA samples and age group (p < 0.001), with most of the PIK3CA exon 9 being ≤ 50 years old (72.4%) and PIK3CA exon 20 being > 50 years old. No statistically significant association was observed between the location of PIK3CA mutation (exons 9 and 20) and the breast site, histopathological diagnosis, and molecular subtypes. Comparisons with existing literature and inconsistencies in PIK3CA mutation frequencies across different BC subtypes underline the need for population-specific research. The study emphasizes the importance of assessing PIK3CA mutations in BC management, offering insights for personalized therapies and potential advancements in understanding this complex disease within the Indonesian context.
Collapse
Affiliation(s)
- Yan Wisnu Prajoko
- Department of Surgical Oncology, Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr. Sardjito General Hospital, Yogyakarta, Indonesia
- Department of Surgery, Division of Cardiac, Thoracic, and Vascular Surgery, Faculty of Medicine, Public Health, and Nursing/Dr. Sardjito General Hospital, Yogyakarta, Indonesia
- Collaboration Research Center for Precision Oncology based Omics- PKR PrOmics, Yogyakarta, Indonesia
| | - Bayu Tirta Dirja
- Department of Microbiology, Faculty of Medicine, Mataram University, Mataram, Indonesia
| | - Susanto Susanto
- Semarang Medical Center (SMC) Telogorejo Hospital, Semarang, Indonesia
| | - Vincent Lau
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Andrew Nobiantoro Gunawan
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Brigitta Natasya Halim
- Department of Anatomical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Nur Dina Amalina
- Department of Pharmaceutical Sciences, Faculty of Medicine, Universitas Negeri Semarang, Semarang, Indonesia
| |
Collapse
|
12
|
Chakravarti B, Tomar MS, Qais FA, Raza S, Abdullah KM, Sharma G, Tewari A, Yadav A, Gupta P, Chattopadhyay N, Shrivastava A, Sinha RA, Siddiqui JA. Alpha lipoic acid modulates metabolic reprogramming in breast cancer stem cells enriched 3D spheroids by targeting phosphoinositide 3-kinase: In silico and in vitro insights. Biomed Pharmacother 2025; 187:118121. [PMID: 40327992 DOI: 10.1016/j.biopha.2025.118121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Breast cancer stem cells (BCSCs) are a unique subpopulation of tumor cells driving tumor resistance, progression, metastasis, and recurrence. Reprogrammed cellular metabolism and key signaling pathways, including Wnt/β-catenin, TGF-β, STAT3, and PI3K/AKT/mTOR pathway play a vital role in maintaining BCSCs. Importantly, PI3K/Akt/mTOR pathway regulates metabolism, survival, growth, and invasion, with PIK3CA, encoding the PI3K catalytic subunit p110α, the most frequently mutated gene in breast cancer. This study investigates the effects of alpha-lipoic acid (LA) on the metabolic profile of BCSCs, focusing on its interaction with PI3K signaling. LA was found to bind PI3K, disrupting cancer-associated metabolic pathways and significantly inhibiting BCSC metabolism. Metabolomic analysis of MCF-7 and MDA-MB-231-derived breast cancer spheroids showed LA-induced metabolic shifts. In MCF-7 spheroids, LA induced upaccumulation of 15 metabolites and downaccumulation of 5, while in MDA-MB-231 spheroids, it induced upaccumulation of 3 and downaccumulation of 16. LA also enhanced the sensitivity of breast cancer spheroids to doxorubicin (Dox), demonstrating a synergistic effect. Mechanistically, LA modulates the PI3K/Akt/mTOR pathway, impairing cell survival and proliferation. These findings highlight the potential of LA as a therapeutic agent for reprogramming cancer metabolism and enhancing chemotherapy efficacy. These results provide a strong rationale for incorporating LA into combination therapy strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Bandana Chakravarti
- Stem Cell/Cell Culture Lab, Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India.
| | - Manendra Singh Tomar
- Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Faizan Abul Qais
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - K M Abdullah
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gunjan Sharma
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Archana Tewari
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Abhishek Yadav
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Pratima Gupta
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Ashutosh Shrivastava
- Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Jawed Akhtar Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
13
|
Saeed MU, Choudhury A, Ansari J, Mohammad T, Hussain A, Fatima U, Alajmi MF, Hassan MI. Discovering potential therapeutic targets in glioblastoma multiforme using a multi-omics approach. Pathol Res Pract 2025; 269:155942. [PMID: 40168776 DOI: 10.1016/j.prp.2025.155942] [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: 10/27/2024] [Revised: 03/08/2025] [Accepted: 03/09/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor associated with high fatality rates, poor prognosis, and limited treatment options. In this study, we utilized RNA-Seq gene count data from GBM patients, sourced from the Gene Expression Omnibus (GEO) database, to conduct an in-depth analysis of gene expression patterns. METHODS Our investigation involved stratifying samples into two distinct sets, Group I and Group II, comparing normal, low-grade, and GBM tumor samples, respectively. Subsequently, we performed differential expression analysis and enrichment analysis to uncover significant gene signatures. To elucidate the protein-protein interactions associated with GBM, we used the STRING plugin within Cytoscape for comprehensive network visualization and analysis. RESULTS By applying Maximal clique centrality (MCC) scores, we identified a set of 10 hub genes in each group. These hub genes were subjected to survival analysis, highlighting their prognostic relevance. In Group I, comprising BUB1, DLGAP5, BUB1B, CDK1, TOP2A, CDC20, KIF20A, ASPM, BIRC5, and CCNB2, these genes emerged as potential biomarkers associated with the transition to low-grade tumors. In Group II, genes such as LIF, LBP, CSF3, IL6, CCL2, SAA1, CCL20, MMP9, CXCL10, and MMP1 were found to be involved in the transformation to adult glioblastoma. Kaplan-Meier's overall survival analysis of these hub genes revealed that modifications, particularly the upregulation of these candidate genes, were associated with reduced survival in GBM patients. CONCLUSIONS The findings established the significance of genomic alterations and differential gene expression in GBM, presenting opportunities for prognostic and targeted therapeutic interventions. This study provides valuable insights into potential avenues for enhancing the clinical management of GBM.
Collapse
Affiliation(s)
- Mohammad Umar Saeed
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Jaoud Ansari
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Urooj Fatima
- UAMS, Donald W. Reynolds Institute on Aging, Department of Geriatrics, Little Rock, AR 72205, USA
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
14
|
Huang J, Yang J, Yang Y, Lu X, Xu J, Lu S, Pan H, Zhou W, Li W, Chen S. Mitigating Doxorubicin-Induced Cardiotoxicity and Enhancing Anti-Tumor Efficacy with a Metformin-Integrated Self-Assembled Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415227. [PMID: 40052211 PMCID: PMC12061326 DOI: 10.1002/advs.202415227] [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] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/06/2025] [Indexed: 05/10/2025]
Abstract
Doxorubicin (Dox) is a potent chemotherapeutic agent commonly used in cancer treatment. However, cardiotoxicity severely limited its clinical application. To address this challenge, a novel self-assembled nanomedicine platform, PMDDH, is developed for the co-delivery of Dox and metformin, an antidiabetic drug with cardioprotective and anti-tumor properties. PMDDH integrates metformin into a polyethyleneimine-based bioactive excipient (PMet), with Dox intercalated into double-stranded DNA and a hyaluronic acid (HA) coating to enhance tumor targeting. The PMDDH significantly improves the pharmacokinetics and tumor-targeting capabilities of Dox, while metformin enhances the drug's anti-tumor activity by downregulating programmed cell death ligand 1 (PD-L1) and activating the AMP-activated protein kinase (AMPK) signaling pathway. Additionally, the DNA component stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which synergizes with Dox-induced immunogenic cell death (ICD) to promote a robust anti-tumor immune response. PMDDH markedly reduces Dox-induced cardiotoxicity by preserving mitochondrial function, reducing reactive oxygen species (ROS) production, and inducing protective autophagy in cardiomyocytes. These findings position PMDDH as a promising dual-function nanomedicine that enhances the anti-tumor efficacy of Dox while minimizing its systemic toxicity, offering a safer and more effective alternative for cancer therapy.
Collapse
Affiliation(s)
- Jiaxin Huang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Jieru Yang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Yuanying Yang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Xiaofeng Lu
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
- Department of CardiologyShanghai General Hospital Jiuquan HospitalNo. 22, West StJiuquanGansu735000China
| | - Juan Xu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| | - Shan Lu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Hong Pan
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical PreparationsSchool of Pharmaceutical ScienceChangsha Medical UniversityChangshaHunan410219China
| | - Wenqun Li
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Songwen Chen
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| |
Collapse
|
15
|
Zhang W, Jia X, Lou D, Wu Q, Yan Y, Mao F. Comparison of mTOR inhibitors combined with endocrine therapy versus that alone in breast cancer: a meta-analysis. Future Oncol 2025; 21:1417-1427. [PMID: 40152674 PMCID: PMC12051556 DOI: 10.1080/14796694.2025.2485022] [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: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND This meta-analysis aims to evaluate the efficacy and safety of rapamycin (mTOR) inhibitors with endocrine therapy versus endocrine therapy alone in treating advanced or metastatic estrogen receptor/progesterone receptor (ER/PR) + breast cancer. METHODS We conducted a comprehensive search in PubMed, Web of Science, Embase, and the Cochrane Library for randomized controlled trials (RCTs) comparing mTOR inhibitors plus endocrine therapy with endocrine therapy alone up to September 2024. RESULTS This analysis included 10 RCTs comprising 3,337 patients. Relative to endocrine therapy alone, the combination of mTOR inhibitors and endocrine therapy significantly improved the clinical benefit rate (RR = 1.41, p < 0.001), overall response rate (RR = 1.40, p = 0.006), progression-free survival (PFS; HR = 0.67, p < 0.001), and overall survival (OS; HR = 0.86, p = 0.056), although the improvement in OS was not statistically significant. Subgroup analyses indicated a more pronounced PFS advantage in patients under 65 years of age (HR = 0.55, p = 0.013) and those who had previously received chemotherapy (HR = 0.51, p = 0.001). However, the incidence of adverse events was higher in the combination therapy group, notably stomatitis (p < 0.001), elevated aspartate aminotransferase/alanine aminotransferase (p = 0.04), and diarrhea (p = 0.01). CONCLUSIONS The combination of mTOR inhibitors with endocrine therapy offers superior efficacy with manageable toxicities in patients with advanced or metastatic ER/PR+ breast cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xinru Jia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Dandi Lou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qingping Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yici Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Feiyan Mao
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
16
|
Arnab SP, Campelo dos Santos AL, Fumagalli M, DeGiorgio M. Efficient Detection and Characterization of Targets of Natural Selection Using Transfer Learning. Mol Biol Evol 2025; 42:msaf094. [PMID: 40341942 PMCID: PMC12062966 DOI: 10.1093/molbev/msaf094] [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: 11/04/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Abstract
Natural selection leaves detectable patterns of altered spatial diversity within genomes, and identifying affected regions is crucial for understanding species evolution. Recently, machine learning approaches applied to raw population genomic data have been developed to uncover these adaptive signatures. Convolutional neural networks (CNNs) are particularly effective for this task, as they handle large data arrays while maintaining element correlations. However, shallow CNNs may miss complex patterns due to their limited capacity, while deep CNNs can capture these patterns but require extensive data and computational power. Transfer learning addresses these challenges by utilizing a deep CNN pretrained on a large dataset as a feature extraction tool for downstream classification and evolutionary parameter prediction. This approach reduces extensive training data generation requirements and computational needs while maintaining high performance. In this study, we developed TrIdent, a tool that uses transfer learning to enhance detection of adaptive genomic regions from image representations of multilocus variation. We evaluated TrIdent across various genetic, demographic, and adaptive settings, in addition to unphased data and other confounding factors. TrIdent demonstrated improved detection of adaptive regions compared to recent methods using similar data representations. We further explored model interpretability through class activation maps and adapted TrIdent to infer selection parameters for identified adaptive candidates. Using whole-genome haplotype data from European and African populations, TrIdent effectively recapitulated known sweep candidates and identified novel cancer, and other disease-associated genes as potential sweeps.
Collapse
Affiliation(s)
- Sandipan Paul Arnab
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | - Matteo Fumagalli
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- The Alan Turing Institute, London, UK
| | - Michael DeGiorgio
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
17
|
Shaw P, Dey Bhowmik A, Gopinatha Pillai MS, Robbins N, Dwivedi SKD, Rao G. Anoikis resistance in Cancer: Mechanisms, therapeutic strategies, potential targets, and models for enhanced understanding. Cancer Lett 2025; 624:217750. [PMID: 40294841 DOI: 10.1016/j.canlet.2025.217750] [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: 01/21/2025] [Revised: 04/01/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Anoikis, defined as programmed cell death triggered by the loss of cell-extracellular matrix (ECM) and cell-cell interactions, is crucial for maintaining tissue homeostasis and preventing aberrant cell migration. Cancer cells, however, display anoikis resistance (AR) which in turn enables cancer metastasis. AR results from alterations in apoptotic signaling, metabolic reprogramming, autophagy modulation, and epigenetic changes, allowing cancer cells to survive in detached conditions. In this review we describe the mechanisms underlying both anoikis and AR, focusing on intrinsic and extrinsic pathways, disrupted cell-ECM interactions, and autophagy in cancer. Recent findings (i.e., between 2014 and 2024) on epigenetic regulation of AR and its role in metastasis are discussed. Therapeutic strategies targeting AR, including chemical inhibitors, are highlighted alongside a network analysis of 122 proteins reported to be associated with AR which identifies 53 hub proteins as potential targets. We also evaluate in vitro and in vivo models for studying AR, emphasizing their role in advancing metastasis research. Our overall goal is to guide future studies and therapeutic developments to counter cancer metastasis.
Collapse
Affiliation(s)
- Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Nathan Robbins
- James E. Hurley School of Science and Mathematics, Oklahoma Baptist University, Shawnee, OK, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA.
| |
Collapse
|
18
|
Wang X, Ren G, Chen B. Integrating metabolomics and network pharmacology to study the mechanism of Er-Xian decoction in improving intervertebral disc degeneration. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119702. [PMID: 40139579 DOI: 10.1016/j.jep.2025.119702] [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: 12/13/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Intervertebral disc degeneration (IDD) is the progressive deterioration of the structure and function of an intervertebral disc (IVD), which manifests as excessive catabolism of the IVD extracellular matrix, which may lead to the gradual loss of IVD proteoglycans and water, thus altering the IVD composition and eventually leading to degeneration. As a traditional Chinese medicine, Er-Xian decoction (EXD) can balance the body's yin and yang, tonify the liver and kidney, invigorate blood circulation, and prevent blood stasis. Pharmacological research has shown that EXD regulates antioxidant and endocrine metabolism, maintains immune balance, and improves microcirculation. AIMS OF THE STUDY To clarify the efficacy of EXD on treating IDD. MATERIALS AND METHODS Serum was collected from model IDD rabbits treated with EXD for metabolomics analysis, and its mechanism of action was predicted on the basis of the metabolomics and network pharmacology data. Nucleus pulposus cells (NPCs) were induced with IL-1β to build an in vitro IDD model, and EXD was administered along with an inhibitor. All groups of cells were subjected to CCK-8 assays, ELISA and flow cytometry, immunohistochemistry, Western blot, and immunofluorescence staining analyses to explore how EXD protects NPCs and the underlying mechanism. RESULTS EXD reduced inflammatory processes, restored IVD height, and alleviated IDD in rabbits. Integrated metabolomics and network pharmacology analyses revealed that EXD exerts its therapeutic effects on IDD primarily via the mTOR and HIF-1 signalling pathways, and the active components of EXD, including anhydroicaritin, β-sitosterol, kaempferol, quercetin, and stigmasterol, bound strongly to pivotal targets within these pathways. Moreover, EXD reduced the inflammatory factor levels, inhibited NPC apoptosis, and upregulated the key proteins p-mTOR, HIF-1α, and p-AKT. Conversely, the HIF-1 inhibitor BAY872243 increased the inflammatory factor levels and led to NPC deterioration. CONCLUSION EXD regulates disc cell metabolism and inflammatory responses by modulating the mTOR and HIF-1 signalling pathways, thereby slowing or reversing IDD.
Collapse
Affiliation(s)
- Xiaobo Wang
- Ningbo Medical Center Lihuili Hospital, 57 Xingning Road, Yinzhou District, Ningbo City, Zhejiang Province, China
| | - Guoqiang Ren
- Ningbo Medical Center Lihuili Hospital, 57 Xingning Road, Yinzhou District, Ningbo City, Zhejiang Province, China
| | - Binhui Chen
- Ningbo Medical Center Lihuili Hospital, 57 Xingning Road, Yinzhou District, Ningbo City, Zhejiang Province, China.
| |
Collapse
|
19
|
Nguyen PT, Tran Huynh QD, Nguyen MD. Isolation and purification of sinensetin, and in silico screening of phytochemicals from Orthosiphon aristatus (Blume) Miq. for anti-breast cancer activity. Nat Prod Res 2025:1-10. [PMID: 40279383 DOI: 10.1080/14786419.2025.2495169] [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: 10/16/2024] [Revised: 03/17/2025] [Accepted: 04/15/2025] [Indexed: 04/27/2025]
Abstract
Orthosiphon aristatus (Blume) Miq. (O. aristatus), a traditional herbal medicine, exhibits a wide range of biological activities but its anti-breast cancer effect is not fully established. In this study, sinensetin was isolated and purified through a multistep process, beginning with liquid-liquid partitioning using dichloromethane, followed by classic column chromatography using gradient elution. Preparative high-performance liquid chromatography with acetonitrile-water (41:59, v/v) mobile phase successfully isolated sinensetin to 95% purity. In silico screening of O. aristatus-derived bioactive compounds revealed that flavonoids (including sinensetin), polyphenols (i.e. 2,3-O-dicaffeoyltartaric acid, rosmarinic acid), diterpenoid (i.e. trans-ozic acid), triterpenoids and dipeptide (i.e. aurantiamide acetate) have good binding affinities with 17β-HSD1, ErbB2/HER2 and PI3K-PKB/Akt proteins. These findings suggest that O. arsistatus could be a rich natural source of inhibitors targeting molecular pathways involved in breast cancer, offering promising therapeutic benefits.
Collapse
Affiliation(s)
- Phuong Tram Nguyen
- Research Group in Pharmaceutical and Biomedical Sciences, Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Quoc-Dung Tran Huynh
- Institute of Pharmaceutical Education and Research, Binh Duong University, Binh Duong, Vietnam
| | - Minh Duc Nguyen
- Research Group in Pharmaceutical and Biomedical Sciences, Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
20
|
Bai J, Gao Y, Zhang G. The treatment of breast cancer in the era of precision medicine. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0510. [PMID: 40269562 PMCID: PMC12032834 DOI: 10.20892/j.issn.2095-3941.2024.0510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
The management of breast cancer, one of the most common and heterogeneous malignancies, has transformed with the advent of precision medicine. This review explores current developments in genetic profiling, molecular diagnostics, and targeted therapies that have revolutionized breast cancer treatment. Key innovations, such as cyclin-dependent kinases 4/6 (CDK4/6) inhibitors, antibody-drug conjugates (ADCs), and immune checkpoint inhibitors (ICIs), have improved outcomes for hormone receptor-positive (HR+), HER2-positive (HER2+), and triple-negative breast cancer (TNBC) subtypes remarkably. Additionally, emerging treatments, such as PI3K inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors, and mRNA-based therapies, offer new avenues for targeting specific genetic mutations and improving treatment response, particularly in difficult-to-treat breast cancer subtypes. The integration of liquid biopsy technologies provides a non-invasive approach for real-time monitoring of tumor evolution and treatment response, thus enabling dynamic adjustments to therapy. Molecular imaging and artificial intelligence (AI) are increasingly crucial in enhancing diagnostic precision, personalizing treatment plans, and predicting therapeutic outcomes. As precision medicine continues to evolve, it has the potential to significantly improve survival rates, decrease recurrence, and enhance quality of life for patients with breast cancer. By combining cutting-edge diagnostics, personalized therapies, and emerging treatments, precision medicine can transform breast cancer care by offering more effective, individualized, and less invasive treatment options.
Collapse
Affiliation(s)
- Jingwen Bai
- The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Yiyang Gao
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
| | - Guojun Zhang
- The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
| |
Collapse
|
21
|
Sarsenbayeva A, Sadak S, Kucuk I, Kudreyeva L, Bakytzhanovna AM, Uslu B. Molybdenum-Based Electrochemical Sensors for Breast Cancer Biomarker Detection: Advances and Challenges. Crit Rev Anal Chem 2025:1-21. [PMID: 40257753 DOI: 10.1080/10408347.2025.2487581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Breast cancer, which is considered the most common type of cancer among women worldwide, is estimated to reach 4.4 million cases in 2070. Early diagnosis has become very important to prevent this expected increase. Various traditional methods, such as mammography, biopsy, enzyme immunoassay (EI), liquid biopsy, immunohistochemistry (IGH), fluorescence in situ hybridization (FISH) are used to diagnose breast cancer, but the fact that these methods are very expensive, have low sensitivity, and cause mutations in tissues due to X-rays has led researchers to discover faster, more cost-effective, and easily detectable methods. In particular, increased levels of new blood-based biomarkers in the circulation can be detected sensitively and selectively by electrochemical methods to facilitate early disease screening and rapid diagnosis. This comprehensive review focuses on the prevalence and pathology of breast cancer, clinical diagnosis of breast cancer, and electrochemical sensors of molybdenum-based compounds for the detection of various breast cancer biomarkers in recent years. Electrochemical analysis studies carried out in the field in recent years are compiled and are considered as aptamer-based, nucleotide-based, and immunosensors. The chemical properties of molybdenum compounds are discussed, and the modifications of these compounds to the electrode surface are discussed under 4 headings: drop casting, electrodeposition, atomic layer deposition, and electrophoretic deposition.
Collapse
Affiliation(s)
- Aliya Sarsenbayeva
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Selenay Sadak
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
- The Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Ipek Kucuk
- The Graduate School of Health Sciences, Ankara University, Ankara, Turkey
- Department of Analytical Chemistry, Faculty of Pharmacy, Başkent University, Ankara, Turkey
| | - Leila Kudreyeva
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Abu Moldir Bakytzhanovna
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Bengi Uslu
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
22
|
Huang B, Yuan Q, Sun J, Wang C, Yang D. Thymidine phosphorylase in nucleotide metabolism: physiological functions and its implications in tumorigenesis and anti-cancer therapy. Front Immunol 2025; 16:1561560. [PMID: 40303404 PMCID: PMC12037492 DOI: 10.3389/fimmu.2025.1561560] [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: 01/16/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Thymidine phosphorylase (TYMP), a protein found in both prokaryotic and eukaryotic cells, is encoded by a gene located in the q13 region of chromosome 22. With a relative molecular mass of 55,000, TYMP exists as a homodimer. Recent research has increasingly illuminated the diverse functions of TYMP. It is known to facilitate platelet activation, osteoclast differentiation, and angiogenesis. Mutations in the TYMP gene are linked to mitochondrial neurogastrointestinal encephalomyopathy. Beyond its physiological roles, TYMP contributes significantly to tumor growth and cancer progression, where it promotes angiogenesis, modulates epigenetic genes, inhibits apoptosis, and acts as a critical enzyme in the nucleoside metabolic rescue pathway. Moreover, TYMP holds substantial implications in cancer treatment and prognosis. Given its involvement in cancer progression, TYMP inhibitors may prove valuable in inhibiting tumor growth and metastasis. Interestingly, while TYMP can drive tumor growth, certain concentrations of TYMP also enhance the cytotoxic effects of chemotherapy drugs such as 5-fluorouracil (5-FU). Although challenges exist-such as the potential disruption of normal physiological functions when inhibiting TYMP-the protein remains a promising target for cancer treatment. Ongoing research on TYMP could deepen our understanding of human physiology and the pathogenesis of cancer and open new avenues for therapeutic interventions. This article provides a comprehensive review of TYMP's structure, physiological functions, and its role in tumorigenesis and anti-tumor therapy.
Collapse
Affiliation(s)
- Bo Huang
- Liaoning Cancer Hospital & Institute, Shenyang, China
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiaao Sun
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chao Wang
- Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Dong Yang
- Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
23
|
Zhao X, Su S, Zhou J, Gao J, Tang X, Wen B. Metabolism and Excretion of 8-O-Acetylharpagide in Rats and Identification of Its Potential Anti-Breast Cancer Active Metabolites. Drug Des Devel Ther 2025; 19:2795-2815. [PMID: 40231196 PMCID: PMC11995923 DOI: 10.2147/dddt.s487898] [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: 07/22/2024] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Background Ajuga decumbens, a traditional Chinese medicine, possesses anti-breast cancer effects. Its main component, 8-O-acetylharpagide, exhibits potential anticancer activity; however, the active metabolites and mechanisms underlying its effects remain unclear. Methods The metabolism and excretion of 8-O-acetylharpagide in rats were investigated using ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry analysis of bile, urine, and feces. Active metabolites were identified and evaluated using network pharmacology, molecular docking, and Western blotting assays. Results A total of 21 metabolites were identified, with demethylation, hydrolysis, and glucuronidation being the primary metabolic pathways. M3 and M5 were identified as key metabolites, showing strong binding affinity to cancer-related targets, such as AKT1, MMP9, and STAT3. M5 displayed strong pharmacokinetic properties, including better lipid solubility and reduced polarity. Network pharmacology analysis indicated that these metabolites exert anticancer effects by modulating the PI3K/AKT signaling pathway. In vivo experiments demonstrated that oral administration of 8-O-acetylharpagide significantly inhibited the proliferation of 4T1 tumor tissues by suppressing the expression of the AKT/NF-κB/MMP9 signaling axis. This may be related to inhibition of the expression of the AKT/NF-κB/MMP9 signaling axis in 4T1 tumor tissues after metabolism of 8-O-acetylharpagide to M5 and M3. Conclusion As a prodrug, 8-O-acetylharpagide is metabolized to M5, which may subsequently exert an anti-breast cancer effect through downregulation of the AKT/NF-κB/MMP9 signaling axis. This study provides a theoretical basis for the clinical application of Ajuga decumbens in breast cancer therapy.
Collapse
MESH Headings
- Animals
- Rats
- Female
- Rats, Sprague-Dawley
- Cell Proliferation/drug effects
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Antineoplastic Agents, Phytogenic/metabolism
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/chemistry
- Mice
- Drug Screening Assays, Antitumor
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/metabolism
- Humans
- Molecular Structure
- Dose-Response Relationship, Drug
- Molecular Docking Simulation
- Administration, Oral
Collapse
Affiliation(s)
- Xinyu Zhao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Sijia Su
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Jingna Zhou
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Junfeng Gao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Xu Tang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Binyu Wen
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| |
Collapse
|
24
|
Zhang S, Lan X, Lei L. LINC01559: roles, mechanisms, and clinical implications in human cancers. Hum Cell 2025; 38:83. [PMID: 40205068 DOI: 10.1007/s13577-025-01218-7] [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: 12/15/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Long intergenic non-protein coding RNA 1559 (LINC01559), a long non-coding RNA (lncRNA) located on chromosome 12p13.1, plays a critical role in the progression of various cancers. The aberrant expression of LINC01559 significantly impacts multiple biological processes in tumor cells, including cell proliferation, epithelial-mesenchymal transition (EMT), migration, invasion, angiogenesis, and cellular stemness. Notably, the expression levels of LINC01559 correlate with the pathological features and prognosis of several cancers, such as pancreatic, breast, and gastric cancers, and it may serve as a diagnostic marker for non-small cell lung cancer. Moreover, the expression of LINC01559 is regulated by various mechanisms and can influence cancer initiation and progression through a competing endogenous RNA (ceRNA) network, where it interacts with a cohort of eight different microRNAs (miRNAs). Additionally, LINC01559 may directly interact with downstream proteins, thereby promoting their functions or enhancing their stability. LINC01559 is also implicated in key signaling pathways associated with cancer development, including the PI3 K/AKT, RAS, and autophagy signaling pathways. Furthermore, it has been linked to drug resistance in breast cancer and hepatocellular carcinoma. This review provides a comprehensive assessment of the clinical implications of dysregulated LINC01559 expression across various cancer types, highlighting its crucial functions and underlying molecular mechanisms in tumorigenesis. Additionally, we present in-depth discussions and propose hypotheses regarding the functional roles of LINC01559 in cancer pathogenesis, while outlining potential research avenues for future exploration of this molecular target.
Collapse
Affiliation(s)
- Shuwen Zhang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Xin Lan
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Ling Lei
- Prevention and Treatment Center, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, 332000, Jiangxi, China.
| |
Collapse
|
25
|
Yin Q, Zhang Y, Xie X, Hou M, Chen X, Ding J. Navigating the future of gastric cancer treatment: a review on the impact of antibody-drug conjugates. Cell Death Discov 2025; 11:144. [PMID: 40188055 PMCID: PMC11972320 DOI: 10.1038/s41420-025-02429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Gastric cancer, marked by its high incidence and poor prognosis, demands the urgent development of novel and effective treatment strategies, especially for patients ineligible for surgery or those who have had limited success with chemotherapy, radiotherapy and targeted therapies. Recently, antibody-drug conjugates (ADCs) have become a key area of investigation due to their high specificity and potent antitumor effects. These therapies combine monoclonal antibodies, designed to bind to tumor-specific antigens, with cytotoxic agents that selectively target and destroy malignant cells. ADCs have generated significant interest in clinical trials as a promising approach to improve both treatment efficacy and patient outcomes in gastric cancer. However, their clinical application is not without challenges and limitations that must be addressed. This review discusses the recent progress in the use of ADCs for gastric cancer treatment.
Collapse
Affiliation(s)
- Qingling Yin
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Yanlong Zhang
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Xueqing Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Meijun Hou
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, 563006, China
| | - Xunsheng Chen
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China
| | - Jie Ding
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China.
| |
Collapse
|
26
|
Jiang S, Li C, Liu D, Zeng F, Wei W, He T, Yang W. Role, mechanisms and effects of Radix Bupleuri in anti‑breast cancer (Review). Oncol Lett 2025; 29:166. [PMID: 39963320 PMCID: PMC11831725 DOI: 10.3892/ol.2025.14912] [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: 10/02/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
The prevalence of breast cancer among women has led to a growing need for innovative anti-breast cancer medications and an in-depth investigation into their molecular mechanisms of action, both of which are essential tactics in clinical intervention. In the clinical practice of Traditional Chinese Medicine, Radix Bupleuri and its active components have shown promise as potential anti-breast cancer agents due to their ability to target multiple pathways, exhibit synergistic effects and reduce toxicity. These compounds are considered to enhance the prognosis of patients with cancer, prolong survival and combat chemotherapy resistance. The present review aimed to delve into the anti-breast cancer properties of Radix Bupleuri and its active ingredients, highlighting their mechanisms, such as inhibition of cell proliferation, promotion of apoptosis, metastasis prevention, microenvironment improvement and synergy with certain chemotherapeutic agents. These findings may provide a scientific rationale for combining Radix Bupleuri and its active components with traditional chemotherapy agents for the management of breast cancer.
Collapse
Affiliation(s)
- Shiting Jiang
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chengxia Li
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Dan Liu
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenli Wei
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tao He
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenli Yang
- Institute for Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
27
|
Xie H, Wu Y, Huang J, Shen Q, Li X, Wang L, Lin J, Chi Z, Ke K, Lin X, Chen R, Liao R, Li Y, Huang N. NK Cell Exosomes Alleviate PD-L1 Expression and Facilitate Tumor Immunity by Repressing PI3K-AKT-mTOR Signaling. Immunol Invest 2025; 54:382-395. [PMID: 39748646 DOI: 10.1080/08820139.2024.2445608] [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] [Indexed: 01/04/2025]
Abstract
BACKGROUND Liver cancer (LC) is a deadly malignancy with limited therapeutic options in recent years. Natural killer cell-derived exosomes (NK-exo), as an important bridge of information transmission between cells, also have a certain killing effect on tumor cells. On this basis, this study investigated the specific regulatory mechanism of NK-exo on LC cells. METHODS NK-exo was collected by differential centrifugation. The diameter and size distribution were characterized by dynamic light scattering (DLS), respectively. Western Blot (WB) assay detected the expression levels of exosome marker protein, PD-L1, and PI3K-AKT-mTOR signal-related proteins. The effect of NK-exo treatment on LC cell viability was measured by the CCK-8. With the use of CFDA·SE, we assessed the proliferation ability of CD8+T cells in direct co-culture with LC cells. The content of cytokines secreted by CD8+T cells in each treatment group was determined by enzyme-linked immunosorbent assay (ELISA) kits. We employed flow cytometry to analyze the expression of PD-L1 protein on the surface of LC cells and CD8 level in mice tumor tissues. RESULTS CCK-8 assay demonstrated that NK-exo repressed the cell viability of LC cells. WB uncovered that the protein expressions of PD-L1, p-AKT, and p-mTOR in NK-exo treated LC cells were decreased, which was returned to the control level after the addition of PI3K agonist. When NK-exo-treated LC cells were directly co-cultivated with CD8+T cells, the proliferation ability and cytokine secretion content of T cells were considerably elevated, and the expression of PD-L1 on LC cell surface was considerably reduced. However, these effects were restored to control levels by PI3K agonists.The in vivo experiments also confirmed that NK-exo could effectively inhibit the progression of LC, and the PI3K agonist could restore this effect to the level of the control group. CONCLUSION This study provided the first evidence that exosomes derived from NK cells inhibited the PI3K-AKT-mTOR signaling pathway in LC cells, and reduced PD-L1 expression, thereby promoting tumor immunity. In comparison to traditional immune checkpoint inhibitors, NK-exo possessed unique mechanisms of action and potential advantages. NK-exo holds the promise of becoming an innovative immunotherapy for the treatment of LC.
Collapse
Affiliation(s)
- Hang Xie
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yujie Wu
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jingyao Huang
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Quan Shen
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoyan Li
- Pathology Department, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lili Wang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Junqing Lin
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhen Chi
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Kun Ke
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xin Lin
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rong Chen
- Fujian Medical University Union Medical College, Fuzhou, China
| | - Rihua Liao
- Radiology Department, The First Hospital Affiliated Longyan, Fujian Medical University, Longyan, China
| | - Yong Li
- Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ning Huang
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
28
|
Li Y, Han Q, Sun Q, Wang X, Ran Y, Ma Y, Lu J, Jin Z, Huang J, Wang Y, Wang J, Chai Y, Li H, Zhang JQ. Discovery of highly potent mTOR inhibitors aimed at suppressing the progression of acute myeloid leukemia. Bioorg Chem 2025; 157:108287. [PMID: 40010131 DOI: 10.1016/j.bioorg.2025.108287] [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: 02/09/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
Acute myeloid leukemia (AML) is a common hematological malignancy with complex etiology; however, current standard chemotherapy regimens for AML show limited efficacy and unsatisfactory tolerability. Herein, a novel class of trisubstituted triazine mTOR inhibitors was designed and synthesized, and the optimal compound, HPT-11, exhibited potent inhibition against mTOR kinase and Molm-13 cell proliferation activities with inhibitory IC50 values of 0.7 and 12 nM, respectively. An antitumor mechanism investigation demonstrated that HPT-11 could potently block the downstream signaling pathway of mTOR and effectively induce apoptosis and autophagy. In addition, in vitro metabolic stability tests further confirmed the stable profiles of HPT-11 in artificial gastrointestinal fluids, rat plasma, and liver microsomes incubating conditions. Overall, our current medicinal chemistry work confirmed that compound HPT-11 is a potent mTOR inhibitor with promising activity in vitro, suggesting its potential as a candidate for further development in the treatment of AML.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Qiu Han
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Qiwen Sun
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Xue Wang
- School of Medicine, Yunnan University, 2 Cuihu North Road, Kunming, 650091, PR China
| | - Yunsheng Ran
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Yifei Ma
- The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
| | - Jiangrong Lu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Ziqi Jin
- School of Medicine, Yunnan University, 2 Cuihu North Road, Kunming, 650091, PR China
| | - Jing Huang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Yujie Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Jianta Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China
| | - Yue'e Chai
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China.
| | - Hongliang Li
- School of Medicine, Yunnan University, 2 Cuihu North Road, Kunming, 650091, PR China.
| | - Ji-Quan Zhang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, P.R. China.
| |
Collapse
|
29
|
Ibrahim MM, Azmi MN, Alhawarri MB, Kamal NNSNM, AbuMahmoud H. Synthesis, characterization and bioactivity of new pyridine-2(H)-one, nicotinonitrile, and furo[2,3-b]pyridine derivatives. Mol Divers 2025; 29:1569-1587. [PMID: 39009909 DOI: 10.1007/s11030-024-10934-5] [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: 05/05/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
Pyridone heterocycles, such as furo[2,3-b]pyridines, have emerged as prominent scaffolds in medicinal chemistry due to their versatile pharmacological properties, including significant anticancer activity. In this study, we successfully synthesized new pyridine-2(H)-one, nicotinonitrile, and furo[2,3-b]pyridine derivatives from chalcones bearing 4-(benzyloxy)phenyl and dichlorothiophenyl subunits to explore their therapeutic potential against breast cancer. By employing a synthetic strategy involving Claisen-Schmidt condensation followed by sequential cyclizations and functional modifications, we synthesized and characterized four compounds (MI-S0, MI-S1, MI-S2, and MI-S3) using various spectroscopic methods, including FT-IR, 1H-NMR, 13C-NMR, DEPT, H,H- and C,H-COSY, and HRMS. The in vitro cytotoxic activity of these compounds was evaluated against two breast cancer cell lines, MCF-7 and MDA-MB-231, and compared with a noncancerous breast cell line, MCF-10A. All compounds exhibited potent cytotoxic activities with minimal selectivity toward normal cells. Molecular docking studies targeting the serine/threonine kinase AKT1, estrogen receptor alpha (ERα), and human epidermal growth factor receptor 2 (HER2) revealed strong binding affinities, suggesting a mechanism involving the disruption of key cellular signaling pathways. These findings underscore the potential of furo[2,3-b]pyridine derivatives as promising candidates for further development into anticancer agents, laying the groundwork for future investigations into their selective therapeutic efficacy and molecular mechanisms of action.
Collapse
Affiliation(s)
- Mohammad M Ibrahim
- Department of Chemistry, Faculty of Science, Al Al-Bayt University, P.O. BOX 130040, Al-Mafraq, 25113, Jordan.
| | - Mohamad Nurul Azmi
- School of Chemical Sciences, Universiti Sains Malaysia, 11800, Minden, Pulau Pinang, Malaysia
| | - Maram B Alhawarri
- Department of Pharmacy, Faculty of Pharmacy, Jadara University, P.O.Box 733, Irbid, 21110, Jordan
| | - Nik Nur Syazni Nik Mohamed Kamal
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200, Kepala Batas, Penang, Malaysia
| | - Hasan AbuMahmoud
- Department of Chemistry, Faculty of Science, Al Al-Bayt University, P.O. BOX 130040, Al-Mafraq, 25113, Jordan
| |
Collapse
|
30
|
Sivakoses A, Marcarian HQ, Arias AM, Lam AR, Ihedioha OC, Santamaria-Barria JA, Gurtner GC, Bothwell ALM. Triple negative breast cancer cells acquire lymphocyte proteins and genomic DNA during trogocytosis with T cells. PeerJ 2025; 13:e19236. [PMID: 40183054 PMCID: PMC11967428 DOI: 10.7717/peerj.19236] [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: 01/29/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Trogocytosis is the process by which a recipient cell siphons small membrane fragments and proteins from a donor cell and can be utilized by cancer cells to avoid immune detection. We observed lymphocyte specific protein expressed by triple negative breast cancer (TNBC) cells via immunofluorescence imaging of patient samples. Image analysis of Cluster of Differentiation 45RA (CD45RA) expression, a naïve T cell specific protein, revealed that all stages of TNBCs express CD45RA. Flow cytometry revealed TNBC cells trogocytose CD45 protein from T cells. We also showed that the acquisition of these lymphoid markers is contact dependent. Confocal and super-resolution imaging further revealed CD45+ spherical structures containing T cell genomic DNA inside TNBC cells after co-culture. Trogocytosis between T cells and TNBC cells altered tumor cell expression of PTPRC, the gene that encodes for CD45. Our results revealed that CD45 is obtained by TNBC cells from T cells via trogocytosis and that TNBC cells express CD45 intracellularly and on the membrane.
Collapse
Affiliation(s)
- Anutr Sivakoses
- Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Haley Q. Marcarian
- Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Anika M. Arias
- Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States
| | - Alice R. Lam
- Department of Biophysics, Stanford University School of Medicine, Palo Alto, California, United States
- Department of Biology, Stanford University, Palo Alto, California, United States
| | - Olivia C. Ihedioha
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | | | - Geoffrey C. Gurtner
- Department of Surgery, University of Arizona, Tucson, Arizona, United States
| | - Alfred L. M. Bothwell
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Department of Immunobiology, Yale University, New Haven, Connecticut, United States
| |
Collapse
|
31
|
Peng J, Liu W, Tian J, Shu Y, Zhao R, Wang Y. Non-coding RNAs as key regulators of epithelial-mesenchymal transition in breast cancer. Front Cell Dev Biol 2025; 13:1544310. [PMID: 40201201 PMCID: PMC11975958 DOI: 10.3389/fcell.2025.1544310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
This study examines the critical role of non-coding RNAs (ncRNAs) in regulating epithelial-mesenchymal transition (EMT) in breast cancer, a prevalent malignancy with significant metastatic potential. EMT, wherein cancer cells acquire mesenchymal traits, is fundamental to metastasis. ncRNAs-such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs)-modulate EMT by influencing gene expression and signaling pathways, affecting cancer cell migration and invasion. This review consolidates recent findings on ncRNA-mediated EMT regulation and explores their diagnostic and therapeutic potential. Specifically, miRNAs inhibit EMT-related transcription factors, while lncRNAs and circRNAs regulate gene expression through interactions with miRNAs, impacting EMT progression. Given the influence of ncRNAs on metastasis and therapeutic resistance, advancing ncRNA-based biomarkers and treatments holds promise for improving breast cancer outcomes.
Collapse
Affiliation(s)
- Jing Peng
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wenhui Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiaju Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuncong Shu
- School of life science, Lanzhou University, Lanzhou, China
| | - Rui Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
32
|
Carvalho E, Canberk S, Schmitt F, Vale N. Molecular Subtypes and Mechanisms of Breast Cancer: Precision Medicine Approaches for Targeted Therapies. Cancers (Basel) 2025; 17:1102. [PMID: 40227634 PMCID: PMC11987866 DOI: 10.3390/cancers17071102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer remains one of the most prevalent diseases worldwide, primarily affecting women. Its heterogeneous nature poses a significant challenge in the development of effective and targeted treatments. Molecular characterization has enabled breast cancer to be classified into four main subtypes: luminal A, luminal B, HER2-positive, and triple-negative breast cancer, based on hormone receptor expression and HER2 status. A deeper understanding of these molecular markers and their associated signaling pathways, such as MAPK and PI3K/AKT, is essential for improving prognosis and optimizing treatment strategies. Currently, several therapeutic agents are utilized in neoadjuvant and adjuvant therapies, often in combination with surgical interventions. However, emerging evidence highlights the growing challenge of drug resistance, which significantly limits the efficacy of existing treatments. Addressing this issue may require innovative approaches, including combination therapies and precision medicine strategies, tailored to the molecular profile of each patient. Therefore, a comprehensive understanding of the pathophysiologic mechanisms driving breast cancer progression and resistance is crucial for the development of advanced targeted therapies with greater precision and efficacy. This review aims to explore recent advancements in molecular research related to breast cancer subtypes and provide a critical analysis of current therapeutic approaches within the framework of precision medicine.
Collapse
Affiliation(s)
- Eduarda Carvalho
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
| | - Sule Canberk
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Fernando Schmitt
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
33
|
Lin J, Yang H, Huang R, Xu T. Discovery of a DNA repair-associated radiosensitivity index for predicting radiotherapy efficacy in breast cancer. Front Oncol 2025; 15:1439516. [PMID: 40201348 PMCID: PMC11975882 DOI: 10.3389/fonc.2025.1439516] [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: 05/28/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Radiotherapy is a cornerstone of breast cancer (BRCA) treatment. Accurately predicting tumor radiosensitivity is critical for optimizing therapeutic outcomes and personalizing treatment strategies. DNA repair pathways are key determinants of radiotherapy response. Thus, we aimed to develop a novel DNA repair-related radiosensitivity model and to identify potential targets for enhancing radiotherapy efficacy. Methods A retrospective study was conducted using data from 942 BRCA patients from TCGA database. A radiosensitivity model, comprising a radiosensitivity index, was developed using LASSO regression analysis. Patients were stratified into radiosensitive (RS) and radioresistant (RR) groups based on their radiosensitivity index (RSI). Associations between the RSI, clinicopathological parameters, and PD-L1 status were analyzed. The CIBERSORT and ESTIMATE algorithms were employed to characterize the immune landscape of the tumor microenvironment. The Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and pRRophetic platform were used to predict treatment responses. Key genes identified in the radiosensitivity model were further validated using in vitro qRT-PCR experiments. Results We successfully constructed a radiosensitivity index incorporating 10 DNA repair-related genes. Patients in the RS group exhibited significantly better prognosis compared to the RR group, but this benefit was limited to those receiving radiotherapy. This survival benefit associated with the radiosensitivity signature was absent in patients who did not receive radiotherapy. The RS group displayed a distinct molecular profile characterized by enrichment of TGF-β signaling and protein secretion pathways, potentially contributing to enhanced radiosensitivity. Furthermore, the RS group exhibited increased infiltration of immune cells. Notably, the RS-PD-L1-high subgroup demonstrated the most favorable survival outcomes and highest immune cell infiltration, highlighting their potential responsiveness to immunotherapy. In addition, the RR group exhibited a distinct profile characterized by enrichment of DNA repair pathways and a heightened sensitivity to CDK and HER2 inhibitors. Conversely, this group displayed resistance to DNA-damaging drugs. These findings were supported by in vitro experiments using MCF-7 and radioresistant MCF-7/IR cell lines, confirming differential expression of key radiosensitivity index genes. Conclusion In conclusion, we established a radiosensitivity model for predicting radiotherapy benefit in breast cancer. Our study reveals a strong association between radiosensitivity, enhanced antitumor immunity, and potential immunotherapy benefit, particularly within the RS-PD-L1-high subgroup.
Collapse
Affiliation(s)
- Jianguang Lin
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hainan Yang
- Department of Ultrasound, First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Rongfu Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Tianwen Xu
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
34
|
Zhang D, Chu Y, Li M, Du L. Isogarcinol Reduces MARS Levels and Deactivates the PI3K/AKT Pathway to Suppress the Malignant Properties of Breast Cancer Cells. Cell Biochem Biophys 2025:10.1007/s12013-025-01727-0. [PMID: 40120049 DOI: 10.1007/s12013-025-01727-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Natural products and their extracts are increasingly considered valuable sources for small-molecule anti-cancer drugs. This study investigates the biological impacts of isogarcinol (ISO) on breast cancer (BC) cells and delves into the underlying mechanisms. In vitro, treatment of ISO at 13 μM substantially reduced the viability, proliferation, and mobility of BC. In vivo, ISO treatment at 5, 10, and 15 mg/kg reduced the tumorigenic activity of MDA-MB-231 cells and decreased the levels of Ki-67 and CD31. ISO exerted tumor suppressive effects by reducing the protein level of methionyl-tRNA synthetase (MARS), as the MARS restoration reversed the trends induced by ISO. Phosphorylation levels of phosphatidyl inositol 3 (PI3K) and protein kinase B (AKT) in BC cells were reduced by ISO but restored by MARS. In the presence of MARS upregulation, further treatment of Alpelisib, a suppressor of the PI3K/AKT pathway, suppressed the malignant properties of BC cells. Collectively, these results demonstrate that ISO curbs the malignant behavior of BC cells by reducing the MARS protein level and deactivating the PI3K/AKT pathway. ISO may be considered a promising regimen for the management of BC.
Collapse
Affiliation(s)
- Dechao Zhang
- Pharmacy Intravenous Admixture Services, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, P.R. China
| | - Yunhai Chu
- Pharmacy Intravenous Admixture Services, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, P.R. China
| | - Meng Li
- Traditional Chinese Medicine Data Research Center, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, P.R. China
| | - Lin Du
- Department of Dermatology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, P.R. China.
| |
Collapse
|
35
|
de Moraes FCA, Sano VKT, Pereira CRM, de Laia EA, Stecca C, Magalhães MCF, Tarantino P. Effects of AKT Inhibitors for PIK3CA/AKT1/PTEN-Altered Advanced or Metastatic Breast Cancer: A Meta-Analysis of Randomized Clinical Trials. Clin Breast Cancer 2025:S1526-8209(25)00079-5. [PMID: 40254500 DOI: 10.1016/j.clbc.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 03/03/2025] [Accepted: 03/16/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE We aimed to answer the following question: How effective is the addition of AKT inhibitors to the treatment of advanced or metastatic breast cancer? METHODS We searched PubMed, Embase and Cochrane for randomized controlled trials (RCTs) that investigated AKT inhibitors for advanced or metastatic BC. We computed hazard-ratios (HRs) for binary endpoints. RESULTS A total of 5 RCTs were included in the meta-analysis, comprising 1,334 patients with BC. The use of AKT inhibitors demonstrated a significant improvement in OS (HR 0.70; 95% CI, 0.58-0.85; P < .001) and PFS (HR 0.6797; 95% CI, 0.5499-0.8403; P < .001) in the overall population. Within the PIK3CA/AKT1/PTEN-altered subgroup (n = 645), the OS rate also significantly favored AKT inhibitors over the control group (HR 0.62; 95% CI, 0.42-0.92; P = .019), as well as PFS (HR 0.5224; 95% CI, 0.3366-0.8105; P = .004). CONCLUSIONS Our findings suggest that the incorporation of AKT inhibitors holds promise for treating patients with advanced or metastatic PIK3CA/AKT1/PTEN-altered BC.
Collapse
Affiliation(s)
| | | | | | | | - Carlos Stecca
- Department of Medical Oncology, Mackenzie Evangelical University Hospital, Curitiba, Paraná, Brazil
| | | | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
36
|
Sun D, Hoffman A, Askarian F, Bjånes E, Lin EX, Varner J, Nizet V. The Role of PI3k-Gamma Modulation in Bacterial Infection: A Review of the Literature and Selected Experimental Observations. Antibiotics (Basel) 2025; 14:315. [PMID: 40149125 PMCID: PMC11939471 DOI: 10.3390/antibiotics14030315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/06/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Phosphoinositide 3-kinase is a potent target for cancer therapy due to its significant role in the regulation of cellular growth and proliferation. Dysregulation of the PI3k signaling cascade can constitutively activate growth pathways to trigger the progression of cancer, resulting in the development of multiple inhibitors as cancer therapeutics. Objectives: The wide array of cells expressing PI3k also include immune cells, and the inhibition of these receptors has shown promise in combating inflammation and infectious disease, a relationship we sought to examine further. Methods: We infected wild-type and PI3kγ knockout murine macrophages as well as PI3kγ inhibitor-treated THP-1 human macrophage-like cells with Staphylococcus aureus and quantified inflammation through gene expression analysis, protein secretion assays, and immunofluorescence imaging. Results: We observed that knockout of PI3kγ in murine macrophages alongside pharmacological inhibition through IPI549 treatment in THP-1 cells led to an NF-κB-driven suppression in transcription and release of inflammatory cytokines upon infection with methicillin-resistant Staphylococcus aureus. We were also able to confirm that this suppression of NF-κB translocation and subsequent decrease in inflammatory cytokine release did not compromise and even slightly boosted the bacterial killing ability. Conclusion: PI3k is primarily targeted for cancer therapies, but further exploration can also be carried out on its potential roles in treating bacterial infection.
Collapse
Affiliation(s)
- Daniel Sun
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA;
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (A.H.); (F.A.); (E.B.); (E.X.L.)
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA;
| | - Alexandria Hoffman
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (A.H.); (F.A.); (E.B.); (E.X.L.)
| | - Fatemeh Askarian
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (A.H.); (F.A.); (E.B.); (E.X.L.)
| | - Elisabet Bjånes
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (A.H.); (F.A.); (E.B.); (E.X.L.)
| | - Eric X. Lin
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (A.H.); (F.A.); (E.B.); (E.X.L.)
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA;
| | - Judith Varner
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA;
- Moores Cancer Center, UC San Diego, La Jolla 92093, USA
| | - Victor Nizet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA;
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (A.H.); (F.A.); (E.B.); (E.X.L.)
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA;
| |
Collapse
|
37
|
Zhang B, Zheng J, Zheng S. Cirsiliol suppresses malignant progression of hepatocellular carcinoma via regulation of glutamine metabolism. Am J Transl Res 2025; 17:2145-2153. [PMID: 40226041 PMCID: PMC11982850 DOI: 10.62347/aoty4308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/06/2024] [Indexed: 04/15/2025]
Abstract
BACKGROUND To investigate the therapeutic potential of cirsiliol in hepatocellular carcinoma (HCC), focusing on its impact on glutamine metabolism. METHODS HCC cell lines HCCLM3 and Huh7 were treated with cirsiliol, and cell viability and proliferation were assessed using CCK-8 assay. Intracellular concentrations of glutamine, α-ketoglutaric acid (α-KG), and adenosine triphosphate (ATP) were measured to evaluate glutamine metabolism. A xenograft tumor model was employed to examine the in vivo effects of cirsiliol. Additionally, network pharmacological analysis was used to identify potential targets of cirsiliol in HCC. Western blotting was conducted to analyze the modulation of the PI3K/AKT signaling pathway by cirsiliol. RESULTS Cirsiliol significantly inhibited HCC cell growth both in vitro and in vivo while reducing levels of glutamine, α-KG, and ATP, indicating suppression of glutamine metabolism. Activation of the PI3K signaling pathway reversed the inhibitory effects of cirsiliol on HCC cell growth and metabolism. CONCLUSION Cirsiliol suppresses glutamine metabolism and inhibits the growth of HCC cells by modulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Bin Zhang
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Haishu District, Ningbo 315000, Zhejiang, China
| | - Jianbo Zheng
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Haishu District, Ningbo 315000, Zhejiang, China
| | - Siming Zheng
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Haishu District, Ningbo 315000, Zhejiang, China
| |
Collapse
|
38
|
Luo H, Sun Y, Xu T. Application status and research progress of targeted therapy drugs for hormone receptor-positive breast cancer. Front Med (Lausanne) 2025; 12:1513836. [PMID: 40134916 PMCID: PMC11933059 DOI: 10.3389/fmed.2025.1513836] [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: 10/19/2024] [Accepted: 02/10/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor in women and the leading cause of cancer-related deaths in women. As one of the most common subtypes of breast cancer, patients with hormone receptor-positive (HR+) breast cancer usually experience disease progression over an extended period of time, triggering the search for therapeutic strategies other than endocrine therapy. In recent years, continuous research on various targets has led to dramatic changes in the treatment of hormone receptor-positive breast cancer patients, resulting in prolonged clinical survival. With the redefinition of human epidermal growth factor-2 (HER2) expression, more precise and individualized treatment is possible. This review comprehensively reviews targeted therapies and critical clinical trials for HR+ breast cancer and tracks the latest advances. It also provides valuable insights into the future direction of targeted therapies.
Collapse
Affiliation(s)
- Han Luo
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Yue Sun
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Tiefeng Xu
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
39
|
Sabit H, Attia MG, Mohamed N, Taha PS, Ahmed N, Osama S, Abdel-Ghany S. Beyond traditional biopsies: the emerging role of ctDNA and MRD on breast cancer diagnosis and treatment. Discov Oncol 2025; 16:271. [PMID: 40050490 PMCID: PMC11885725 DOI: 10.1007/s12672-025-01940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Breast cancer management has traditionally relied on tissue biopsies and imaging, which offer limited insights into the disease. However, the discovery of circulating tumor DNA (ctDNA) and minimal residual disease (MRD) detection has revolutionized our approach to breast cancer. ctDNA, which is fragmented tumor DNA found in the bloodstream, provides a minimally invasive way to understand the tumor's genomic landscape, revealing heterogeneity and critical mutations that biopsies may miss. MRD, which indicates cancer cells that remain after treatment, can now be detected using ctDNA and other advanced methods, improving our ability to predict disease recurrence. This allows for personalized adjuvant therapies based on individual MRD levels, avoiding unnecessary treatments for patients with low MRD. This review discusses how ctDNA and MRD represent a paradigm shift towards personalized, genomically guided cancer care, which has the potential to significantly improve patient outcomes in breast cancer.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt.
| | - Manar G Attia
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nouran Mohamed
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Pancé S Taha
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nehal Ahmed
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Salma Osama
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| |
Collapse
|
40
|
Arnab SP, Dos Santos ALC, Fumagalli M, DeGiorgio M. Efficient detection and characterization of targets of natural selection using transfer learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641710. [PMID: 40093065 PMCID: PMC11908262 DOI: 10.1101/2025.03.05.641710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Natural selection leaves detectable patterns of altered spatial diversity within genomes, and identifying affected regions is crucial for understanding species evolution. Recently, machine learning approaches applied to raw population genomic data have been developed to uncover these adaptive signatures. Convolutional neural networks (CNNs) are particularly effective for this task, as they handle large data arrays while maintaining element correlations. However, shallow CNNs may miss complex patterns due to their limited capacity, while deep CNNs can capture these patterns but require extensive data and computational power. Transfer learning addresses these challenges by utilizing a deep CNN pre-trained on a large dataset as a feature extraction tool for downstream classification and evolutionary parameter prediction. This approach reduces extensive training data generation requirements and computational needs while maintaining high performance. In this study, we developed TrIdent, a tool that uses transfer learning to enhance detection of adaptive genomic regions from image representations of multilocus variation. We evaluated TrIdent across various genetic, demographic, and adaptive settings, in addition to unphased data and other confounding factors. TrIdent demonstrated improved detection of adaptive regions compared to recent methods using similar data representations. We further explored model interpretability through class activation maps and adapted TrIdent to infer selection parameters for identified adaptive candidates. Using whole-genome haplotype data from European and African populations, TrIdent effectively recapitulated known sweep candidates and identified novel cancer, and other disease-associated genes as potential sweeps.
Collapse
Affiliation(s)
- Sandipan Paul Arnab
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| | | | - Matteo Fumagalli
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- The Alan Turing Institute, London, UK
| | - Michael DeGiorgio
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
41
|
Ismail DF, El-Keey MM, Elgendy SM, Hessien M. Impregnation of mesenchymal stem cell conditioned media with wortmannin enhanced its antiproliferative effect in breast cancer cells via PI3K/Akt/mTOR pathway. BMC Res Notes 2025; 18:93. [PMID: 40038752 PMCID: PMC11877855 DOI: 10.1186/s13104-025-07124-3] [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: 03/26/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND/AIM Conditioned media derived from Mesenchymal stem cells (MSC-CM) was suggested as a promising alternative cell-free regenerative therapy. It is hypothesized that the synergistic effect of MSC-CM with anticancer drugs may improve their antiproliferative and antimetastatic effects against cancer cells. Herein, the MSC-CM was impregnated with Wortmannin, a pan-PI3K/Akt/mTOR inhibitor, and their combined effect was investigated against breast cancer cells. MATERIALS AND METHODS To explore this, the cytotoxic, apoptotic, and autophagic potentials were assessed in luminal-A breast cancer cells (MCF-7). RESULTS We found that incubation of MCF-7 to Wort-containing-CM induced apoptosis- and autophagy-mediated cell death, meanwhile prolonged exposure caused massive necrotic cell death. The involvement of MSC-CM effectively reduced Wortmannin IC50 observed in Wort-treated cells. Also, Wort-loaded-CM induced nuclear DNA fragmentation and reduced in vitro cell migration. These findings were associated with a Wort-dependent reduction in cell viability, the formation of the phosphorylated Akt and mTOR proteins, reduced the expression of mRNA, and downregulated the expression of the catalytic domain of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K-Ca). CONCLUSION These findings revealed the promising antiproliferative and antimetastasis effects of combining pan-PI3K/Akt/mTOR inhibitors with MSC-derived-CM in breast cancer via the downregulation of PI3K/AKT/mTOR signaling pathways. Further studies are required to validate this chem-regenerative strategy in cancer treatment.
Collapse
Affiliation(s)
- Doha F Ismail
- Molecular Cell Biology Unit, Division of Biochemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mai M El-Keey
- Molecular Cell Biology Unit, Division of Biochemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Saad M Elgendy
- Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
42
|
Khan Y, Rizvi S, Raza A, Khan A, Hussain S, Khan NU, Alshammari SO, Alshammari QA, Alshammari A, Ellakwa DES. Tailored therapies for triple-negative breast cancer: current landscape and future perceptions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03896-4. [PMID: 40029385 DOI: 10.1007/s00210-025-03896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
Triple-negative breast cancer (TNBC) has become one of the most challenging cancers to date due to its great variability in biological features, high growth rate, and rare options for treatment. This review examines several innovative strategies for tailored treatment of TNBC, focusing mainly on the most recent developments and potential directions. The molecular landscape of TNBC is covered in the first section, which keeps the focus on transcriptome and genomic profiling while highlighting key molecular targets like mutations in the BRCA1/2, PIK3CA, androgen receptors (AR), epidermal growth factor receptors (EGFR), and immunological checkpoint molecules. This review also covers novel therapies that aim to block well-defined pathways, including immune checkpoint inhibitors (ICI), EGFR inhibitors, drugs that target AR, poly ADP ribose polymerase (PARP) inhibitors, and drugs that disrupt the PI3K/AKT/mTOR pathway. Additionally, it covers novel strategies focusing on combination therapy, targeting the DNA damage response pathway, and epigenetic modulators. Conclusively, it emphasizes perspectives and directions on topics such as personalized medicine, artificial intelligence (AI), predictive biomarkers, and treatment planning with the inclusion of machine learning (ML).
Collapse
Affiliation(s)
- Yumna Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, 25130, Pakistan.
| | - Sana Rizvi
- Bakhtawar Amin Medical and Dental College, Bakhtawar Amin Trust Teaching Hospital, Multan, Pakistan
| | - Ali Raza
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Amna Khan
- Abbottabad International Medical Institute, Abbottabad, 22020, Pakistan
| | - Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, College of Pharmacy, Northern Border University, 76321, Rafha, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Abdulkarim Alshammari
- Department of Clinical Practice, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Doha El-Sayed Ellakwa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo, Egypt.
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt.
| |
Collapse
|
43
|
Jin C, Gao X, Ni J, Zhang B, Wang Z. MiR-592 Attenuates Tamoxifen Resistance in Breast Cancer Through PIK3CA-Mediated PI3K/AKT/mTOR Signaling Pathway. Appl Biochem Biotechnol 2025; 197:2051-2065. [PMID: 39661080 DOI: 10.1007/s12010-024-05123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
Tamoxifen (TAM) is vital in breast cancer (BC) treatment, yet its resistance significantly impairs its efficacy. While miR-592 is known for its suppressive role in BC, its effect on chemotherapy resistance remains unclear. In this study, we observed a significant reduction in miR-592 levels in TAM-resistant BC tissues and cell lines. Low miR-592 expression was significantly associated with advanced TNM stage, lymph node metastasis, and poorer patient survival. Dual-luciferase assay confirmed miR-592 binding to the predicted gene PIK3CA. Increasing miR-592 levels decreased the IC50 of TAM, inhibited cell viability, migration, and invasion, and enhanced apoptosis in vitro, which was mimicked by PIK3CA knockdown and reversed by PIK3CA overexpression. Moreover, miR-592 upregulation suppressed tumor growth and improved TAM responsiveness in vivo. Molecularly, both si-PIK3CA and miR-592 mimics decreased the expression ratios of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR, while increasing cleaved caspase-3 and E-cadherin expression in MCF-7/TAM cells. PIK3CA overexpression partially reversed these reductions. In conclusion, our study demonstrates that miR-592 attenuates TAM resistance by inhibiting the PIK3CA-driven PI3K/AKT/mTOR signaling pathway, representing a promising strategy to address chemoresistance in BC.
Collapse
Affiliation(s)
- Conghui Jin
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, P.R. China
- Department of Medical Oncology, Nantong University Affiliated Tumor Hospital, Nantong, 226361, Jiangsu, P.R. China
| | - Xiangxiang Gao
- Department of Medical Oncology, Nantong University Affiliated Tumor Hospital, Nantong, 226361, Jiangsu, P.R. China
| | - Jingyi Ni
- Department of Medical Oncology, Nantong University Affiliated Tumor Hospital, Nantong, 226361, Jiangsu, P.R. China
| | - Baochun Zhang
- Department of Medical Oncology, Nantong University Affiliated Tumor Hospital, Nantong, 226361, Jiangsu, P.R. China
| | - Zhenxin Wang
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, P.R. China.
| |
Collapse
|
44
|
Yi S, Qu T, Wu H, Xu C, Xu J, Yu F, Ye L. Knockdown of PLOD2 inhibits pulmonary artery smooth muscle cell glycolysis under chronic intermittent hypoxia via PI3K/AKT signal. Exp Cell Res 2025; 446:114453. [PMID: 39961468 DOI: 10.1016/j.yexcr.2025.114453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
OBJECTIVE This study aimed to investigate the role and potential mechanism of procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) in chronic intermittent hypoxia (CIH)-induced mice and pulmonary arterial smooth muscle cells (PASMCs). METHODS CIH mouse model was pre-injected with AAV-shPLOD2 by tail vein, and the pathological changes of lung was evaluated using hematoxylin-eosin (H&E) and α-SMA immunostaining. Enriched KEGG pathway analyses of PLOD2 targeted genes were performed using GSE11341 and GSE131425 datasets. Next, primary PASMCs were exposed to CIH environment, and then measured its proliferation, migration and glycolysis by CCK8, EdU assay, wound healing assay, Transwell and western blotting. RESULTS PLOD2 expression was increased in the lungs of CIH-induced mice and in PASMCs under CIH conditions. Moreover, glycolysis and PI3K/AKT pathway were regulated by PLOD2. Silencing of PLOD2 significantly inhibited the increase of RV/(LV + S) and RVSP, alleviated pathological changes of lung in CIH-induced mice and restrained the proliferation, migration, glycolysis and activation of PI3K/AKT in CIH-induced PASMCs. The inhibitory effects of PLOD2 silencing on PASMC proliferation and migration were accelerated by 2-DG (an inhibitor of glycolysis) and were reversed by lactate (the end product of glycolysis). In addition, the inhibitory effects of PLOD2 silencing on PASMC proliferation, migration and glycolysis were accelerated by PI3K/AKT inhibitor LY294002 and were reversed by the agonist 740Y-P. CONCLUSIONS Silencing of PLOD2 inhibits PI3K/AKT signaling to limit PASMC glycolysis which allows PASMC proliferation and migration in CIH-induced pulmonary arterial hypertension (PAH).
Collapse
Affiliation(s)
- Shenwen Yi
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Tiange Qu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Heling Wu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Chenyu Xu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Jun Xu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Fei Yu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Liang Ye
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
45
|
Di-Luoffo M, Schmitter C, Barrere EC, Therville N, Chaouki M, D'Angelo R, Arcucci S, Thibault B, Delarue M, Guillermet-Guibert J. Mechanical compressive forces increase PI3K output signaling in breast and pancreatic cancer cells. Life Sci Alliance 2025; 8:e202402854. [PMID: 39746759 PMCID: PMC11707390 DOI: 10.26508/lsa.202402854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Mechanical stresses, including compression, arise during cancer progression. In solid cancer, especially breast and pancreatic cancers, the rapid tumor growth and the environment remodeling explain their high intensity of compressive forces. However, the sensitivity of compressed cells to targeted therapies remains poorly known. In breast and pancreatic cancer cells, pharmacological PI3K inactivation decreased cell number and induced apoptosis. These effects were accentuated when we applied 2D compression forces in mechanically responsive cells. Compression selectively induced the overexpression of PI3K isoforms and PI3K/AKT pathway activation. Furthermore, transcriptional effects of PI3K inhibition and compression converged to control the expression of an autophagy regulator, GABARAP, whose level was inversely associated with PI3K inhibitor sensitivity under compression. Compression alone blocked autophagy flux in all tested cells, whereas inactivation of basal PI3K activity restored autophagy flux only in mechanically non-responsive compressed cells. This study provides direct evidence for the role of the PI3K/AKT pathway in compression-induced mechanotransduction. PI3K inhibition promotes apoptosis or autophagy, explaining PI3K importance to control cancer cell survival under compression.
Collapse
Affiliation(s)
- Mickaël Di-Luoffo
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Céline Schmitter
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Emma C Barrere
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Nicole Therville
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Maria Chaouki
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Romina D'Angelo
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Silvia Arcucci
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Benoit Thibault
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Morgan Delarue
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Julie Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| |
Collapse
|
46
|
Yang Z, Zheng Y, Ren K, Wang W, Li S. Hydroxy-selenomethionine helps cows to overcome heat stress by enhancing antioxidant capacity and alleviating blood-milk barrier damage. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:171-181. [PMID: 39967694 PMCID: PMC11833791 DOI: 10.1016/j.aninu.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/23/2024] [Accepted: 10/30/2024] [Indexed: 02/20/2025]
Abstract
Heat stress can lead to decreased feed intake, apoptosis of mammary epithelial cells, and decreased milk yield and quality. Selenium is an important element in the composition of at least 25 selenoproteins. Hydroxy-selenomethionine (HMSeBA) is a novel organic selenium that has been shown to have a better deposition effect. However, whether HMSeBA alleviates damage to the mammary gland blood-milk barrier caused by heat stress and how this affects the performance of dairy cows remain largely unexplored. Therefore, 32 healthy Holstein cows with similar gestation days (150.41 ± 20.07 d), milk yield (36.15 ± 3.02 kg) and parity (3.25 ± 0.51) were selected and randomly divided into two total mixed rations with different selenium (Se) sources: sodium selenite (SSe) and HMSeBA. This study evaluated the outcomes of HMSeBA on antioxidant capacity, immunity, and blood-milk barrier damage in dairy cows during heat stress by collecting the samples of blood, rumen fluid and mammary gland biopsy. The experiment was conducted over 35 d, including a 5-day pre-feeding period and a 30-day experimental period. The temperature and humidity index (THI) were all above 80 throughout the experiment period. The results showed that HMSeBA decreased the respiratory rate (P < 0.001) and the content of inflammatory cytokines in the serum and increased the content of immune factors and antioxidant capacity (P < 0.05). In addition, HMSeBA reduced the expression of inflammatory cytokines and heat shock proteins in mammary gland (P < 0.05). Hematoxylin-eosin-stained pathological sections showed massive thickening of acinar walls and severe destruction of glandular structures in the SSe group, but the structure of the acinar mammary gland in the HMSeBA group was intact. Furthermore, HMSeBA promoted the expression of the phosphatidylinositol 3-kinase (PI3K, P < 0.001)/protein kinase B (AKT, P = 0.011)/mammalian target of rapamycin (mTOR, P = 0.008) pathway and improved the expression of zonula occludens-1 (ZO-1, P = 0.014) and occluding (OCLN, P = 0.012) in the mammary gland, suggesting less damage caused by heat stress to the blood-milk barrier. Our results demonstrated that HMSeBA can improve the antioxidant capacity and immunity of dairy cows and the expression of tight junction proteins in mammary gland to help alleviate the blood-milk barrier damage by heat stress, which could reduce the damage of heat stress on milk yield.
Collapse
Affiliation(s)
- Zhantao Yang
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuhui Zheng
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Kai Ren
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
47
|
Bahrami N, Abdi M. Knockout of histone deacetylase 8 gene in breast cancer cells may alter the expression pattern of the signaling molecules. Adv Med Sci 2025; 70:27-32. [PMID: 39437892 DOI: 10.1016/j.advms.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/26/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE Breast cancer (BC) is the most common cancer diagnosed in the world and it is also the main leading cause of cancer deaths in women. Change in epigenetic mechanisms promotes BC initiation and progression. Histone deacetylase 8 (HDAC8) was found to act as a potential oncogene in different malignancies. For better understanding of the HDAC8 function in BC development, we investigated the effect of HDAC8 deletion on the expression of genes involved in signaling pathways. MATERIALS AND METHODS In this study, CRISPR technology was used to knockout the HDAC8 gene in MDA-MB-468, MDA-MB-231 and MCF-7 cell lines. For this purpose, two gRNAs were designed and cloned into the PX459 vector. The gRNA-containing vectors were transfected into the BC cell lines and then the effect of this deletion on the expression of genes involved in signaling pathway was determined using quantitative real-time PCR (qRT-PCR). RESULTS Analysis of qRT-PCR results showed a reduction in the expression of studied genes in BC cell lines after deletion of the HDAC8 gene compared to untreated controls. Although this decline was not significant for FGF2 and FGFR1 genes, however the mTOR, IGF1R, INSR, VEGFA and VEGFR2 genes showed statistically significant reduction in the studied BC cell lines. In addition, the down-regulation of PDGFC and PDGFRA genes were only significant in the TNBC cell lines. CONCLUSION Overall, our study showed that HDAC8 can exert its oncogenic effects by altering the expression level of molecules involved in some signaling pathways, and inhibiting HDAC8 can revert these effects.
Collapse
Affiliation(s)
- Nahid Bahrami
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Abdi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
48
|
Fusco N, Malapelle U. Next-generation sequencing for PTEN testing in HR+/HER2- metastatic breast cancer. Crit Rev Oncol Hematol 2025; 207:104626. [PMID: 39909182 DOI: 10.1016/j.critrevonc.2025.104626] [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: 12/18/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
Molecular alterations in the Phosphoinositide 3-kinase (PI3K) pathway are key drivers of tumorigenesis and progression in hormone receptor-positive, HER2-negative (HR+/HER2 -) metastatic breast cancer (MBC). These genomic changes are actionable through targeted therapeutic agents. In particular, access to these therapies depends on accurate molecular testing of PIK3CA, AKT1, and PTEN. Next-generation sequencing (NGS) has emerged as a transformative diagnostic tool, offering a comprehensive analysis of PI3K pathway alterations while concurrently evaluating other actionable markers, such as ESR1 and BRCA. Acknowledging its clinical importance, the European Society for Medical Oncology (ESMO) recommends NGS of tumor or plasma samples as the standard of care for patients with HR+ /HER2 - MBC. Although resource-intensive, NGS represents a significant advancement in MBC diagnostics, ensuring that therapeutic decisions are informed by a detailed and multidimensional molecular profile. This review highlights the capabilities of NGS for PI3K pathway testing in HR+ /HER2 - MBC, with a particular focus on the spectrum of PTEN alterations.
Collapse
Affiliation(s)
- Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Naples 80131, Italy.
| |
Collapse
|
49
|
Sun P, Liu F, Huo K, Wang J, Cheng Y, Shang S, Ma W, Yu J, Han J. Adiponectin facilitates the cell cycle, inhibits cell apoptosis and induces temozolomide resistance in glioblastoma via the Akt/mTOR pathway. Oncol Lett 2025; 29:127. [PMID: 39807099 PMCID: PMC11726000 DOI: 10.3892/ol.2025.14875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/22/2024] [Indexed: 01/16/2025] Open
Abstract
Adiponectin (ADN) regulates DNA synthesis, cell apoptosis and cell cycle to participate in the pathology and progression of glioblastoma. The present study aimed to further explore the effect of ADN on temozolomide (TMZ) resistance in glioblastoma and the underlying mechanism of action. Glioblastoma cell lines (U251 and U87-MG cells) were treated with ADN and TMZ at different concentrations; subsequently, 3.0 µg/ml ADN and 1.0 mM TMZ were selected as the optimal concentrations for the experimental conditions. LY294002 (a PI3K inhibitor) was added to ADN or ADN + TMZ-treated glioblastoma cell lines. Cell growth rate was determined using the Cell Counting Kit-8 assay, the apoptotic rate and cell cycle were evaluated using Annexin V/propidium iodide and cell cycle assays, and p-Akt (Thr308), p-Akt (Ser473), Akt, p-mTOR, c-caspase 3, caspase 3, Bax, cyclin B1 and cyclin D1 expression was determined by western blotting. Adiponectin receptor (ADIPOR) 1 and ADIPOR2 were expressed in glioblastoma cell lines. The glioblastoma cell line growth rate was increased by ADN in a concentration- and time-dependent manner. ADN inhibited glioblastoma cell line apoptosis and facilitated cell cycle. Of note, ADN activated the Akt/mTOR pathway and the addition of LY294002 reversed the effect of ADN, indicating that ADN activated the Akt/mTOR pathway to suppress apoptosis and promote cell cycle in glioblastoma cell lines. Notably, TMZ inhibited glioblastoma cell line growth, promoted apoptosis and increased G2 phase cell cycle arrest. However, the addition of ADN reversed the effect of TMZ in glioblastoma cell lines, disclosing that ADN induced TMZ resistance. Markedly, ADN-mediated TMZ resistance was further attenuated by LY294002, suggesting that ADN activated the Akt/mTOR pathway to induce TMZ resistance in glioblastoma cell lines. In conclusion, ADN activated the Akt/mTOR pathway to facilitate cell cycle, inhibit cell apoptosis and induce TMZ resistance in glioblastoma.
Collapse
Affiliation(s)
- Peng Sun
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fude Liu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianyi Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yawen Cheng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenlong Ma
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jia Yu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianfeng Han
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
50
|
Weng J, Shan Y, Chang Q, Cao C, Liu X. Research progress on N 6-Methyladenosine modification in angiogenesis, vasculogenic mimicry, and therapeutic implications in breast cancer. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:57-70. [PMID: 39710080 DOI: 10.1016/j.pbiomolbio.2024.12.003] [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: 10/07/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
N6-methyladenosine (m6A) modification is the most common epitranscriptomic modification in eukaryotic RNA and has garnered extensive attention in the context of breast cancer research. The m6A modification significantly impacts tumorigenesis and tumor progression by regulating RNA stability, splicing, translation, and degradation. In this review we summarize recent advances in understanding the roles of m6A modification in the mechanisms underlying angiogenesis and vasculogenic mimicry in breast cancer. We review how m6A modification and associated transcripts influence relevant factors by affecting key factors and signaling pathways, highlighting the interactions among m6A "writers," "erasers," and "readers," and their overall impact on tumor angiogenesis and vasculogenic mimicry, as well as potential new therapeutic targets.
Collapse
Affiliation(s)
- Jiachen Weng
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Yisi Shan
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Qingyu Chang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Chenyan Cao
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Xuemin Liu
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China.
| |
Collapse
|