1
|
Chriqui LE, Cavin S, Perentes JY. Dual implication of endothelial adhesion molecules in tumor progression and cancer immunity. Cell Adh Migr 2025; 19:2472308. [PMID: 40071851 PMCID: PMC11913389 DOI: 10.1080/19336918.2025.2472308] [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: 02/15/2024] [Revised: 10/16/2024] [Accepted: 01/19/2025] [Indexed: 03/19/2025] Open
Abstract
Adhesion molecules are proteins expressed at the surface of various cell types. Their main contribution to immunity is to allow the infiltration of immune cells in an inflamed site. In cancer, adhesion molecules have been shown to promote tumor dissemination favoring the development of metastasis. While adhesion molecule inhibition approaches were unsuccessful for cancer control, their importance for the generation of an immune response alone or in combination with immunotherapies has gained interest over the past years. Currently, the balance of adhesion molecules for tumor promotion/inhibition is unclear. Here we review the role of selectins, intercellular adhesion molecules (ICAM) and vascular cell adhesion molecules (VCAM) from the perspective of the dual contribution of adhesion molecules in tumor progression and immunity.
Collapse
Affiliation(s)
- Louis-Emmanuel Chriqui
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| | - Sabrina Cavin
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| | - Jean Yannis Perentes
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [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: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
3
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [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: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
4
|
Rybarczyk A, Sultan T, Hussain N, Azam HMH, Rafique S, Zdarta J, Jesionowski T. Fusion of enzymatic proteins: Enhancing biological activities and facilitating biological modifications. Adv Colloid Interface Sci 2025; 340:103473. [PMID: 40086016 DOI: 10.1016/j.cis.2025.103473] [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: 09/08/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
The fusion of enzymatic proteins represents a dynamic frontier in biotechnology and enzymatic engineering. This in-depth review looks at the many different ways that fusion proteins can be used, showing their importance in biosensing, gene therapy, targeted drug delivery, and biocatalysis. Fusion proteins have shown an astounding ability to improve and fine-tune biological functions by combining the most beneficial parts of different enzymes. Our first step is to explain how protein fusion increases biological functions. This will provide a broad picture of how this phenomenon has changed many fields. We dissect the intricate mechanisms through which fusion proteins orchestrate cellular processes, underscoring their potential to revolutionize the landscape of molecular biology. We also explore the complicated world of structural analysis and design strategies, stressing the importance of molecular insights for making the fusion protein approach work better. These insights broaden understanding of the underlying principles and illuminate the path toward unlocking untapped potential. The review also introduces cutting-edge techniques for constructing fusion protein libraries, such as DNA shuffling and phage display. These new methods allow scientists to build a molecular orchestra with an unprecedented level of accuracy, and thus use fusion proteins to their full potential in various situations. In conclusion, we provide a glimpse into the current challenges and prospects in fusion protein research, shedding light on recent advancements that promise to reshape the future of biotechnology. As we make this interesting journey through the field of enzymatic protein combination, it becomes clear that the fusion paradigm is about to start a new era of innovation that will push the limits of what is possible in biology and molecular engineering.
Collapse
Affiliation(s)
- Agnieszka Rybarczyk
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland
| | - Talha Sultan
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Hafiz Muhammad Husnain Azam
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Universitätsplatz 1, 01968 Senftenberg, Germany
| | - Safa Rafique
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Jakub Zdarta
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland.
| | - Teofil Jesionowski
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland.
| |
Collapse
|
5
|
Villa de la Torre F, Tec Caamal EA, Rizo AM, Argáez RB, Pintor Romero VG, Yáñez-Barrientos E, Morales-Tirado DJ, Alonso-Castro AJ, Argáez VA. In vivo and in vitro anti-inflammatory activity of the methanolic leaves extract of Gymnopodium floribundum Rolfe. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119743. [PMID: 40204249 DOI: 10.1016/j.jep.2025.119743] [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: 10/28/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gymnopodium floribundum Rolfe, known locally as "Dzidzilche" or "Ts'its'ilche," is a native species from Mexico and Central America. In Mayan communities, this plant is used to relieve inflammation and diverse respiratory diseases such as colds, catarrh, bronchitis, and asthma. Usually, a decoction of leaves or flowers is prepared and administered orally. AIM This research explores the anti-inflammatory effects of the methanol extract of Gymnopodium floribundum Rolfe leaves (MGF) using in vitro and in vivo animal models of inflammation. METHODS MGF was characterized by GC-MS, and cytotoxicity was assessed using hemolysis and MTT assays. The antiphlogistic effect in vitro was measuring the release of cytokines, hydrogen peroxide, and nitric oxide in macrophages stimulated with LPS. Additionally, anti- and pro-inflammatory cytokines, prostaglandins, and leukotrienes in serum were quantified in carrageenan-induced mouse paw edema. Finally, 1-fluoro-2,4-dinitrobenzene (DNFB)-induced delayed-type hypersensitivity and TPA-induced ear edema models were analyzed. RESULTS Compounds found in MGF, such as D-pinitol and protocatechuic (3,4-dihydroxybenzoic) acid, are reported to exert anti-inflammatory effects. MGF showed no hemolytic or cytotoxic effects. Nevertheless, it displayed in vitro anti-inflammatory activity by decreasing the release of IL-6, IL-1β, TNF-α, hydrogen peroxide, and nitric oxide levels; on the other hand, it increased IL-10 production. Furthermore, the MGF significantly reduced inflammation in mouse models and reduced the release of leukotrienes, prostaglandins, and pro-inflammatory cytokines. CONCLUSION Gymnopodium floribundum Rolfe exhibits anti-inflammatory activity by suppressing pro-inflammatory mediators, altering cell migration mechanisms, and raising IL-10 production.
Collapse
Affiliation(s)
- Fabiola Villa de la Torre
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| | | | - Abril Martínez Rizo
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| | - Rocío Borges Argáez
- Biotechnology Unit, Scientific Research Center of Yucatan (CICY), Yucatan, Mexico.
| | | | - Eunice Yáñez-Barrientos
- Division of Natural and Exact Sciences, Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | | | - Angel Josabad Alonso-Castro
- Division of Natural and Exact Sciences, Department of Chemistry, University of Guanajuato, Guanajuato, Mexico.
| | - Víctor Arana Argáez
- Faculty of Chemistry, Laboratory of Pharmacology, Autonomous University of Yucatan, Yucatan, Mexico.
| |
Collapse
|
6
|
Wang Z, Zhang J, Luo L, Zhang C, Huang X, Liu S, Chen H, Miao W. Nucleoporin 93 Regulates Cancer Cell Growth and Stemness in Bladder Cancer via Wnt/β-Catenin Signaling. Mol Biotechnol 2025; 67:2072-2084. [PMID: 38744786 DOI: 10.1007/s12033-024-01184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024]
Abstract
Bladder cancer (BLCA) is a prevalent cancer type with an unmet need for new therapeutic strategies. Nucleoporin 93 (Nup93) is implicated in the pathophysiology of several cancers, but its relationship with bladder cancer remains unclear. Nup93 expression was analyzed in TCGA datasets and 88 BLCA patient samples. Survival analysis and Cox regression models evaluated the association between Nup93 levels and patient prognosis. BLCA cells were used to investigate the effects of Nup93 overexpression or knockdown on cell growth, invasion, stemness (sphere formation and ALDH2 + cancer stem cell marker), and Wnt/β-catenin signaling in vitro. The Wnt activator BML-284 was used to confirm the involvement of Wnt/β-catenin signaling pathway. A xenograft mouse model validated the in vitro findings. Nup93 was highly expressed in BLCA tissues and cell lines, and high Nup93 expression correlated with poor prognosis in BLCA patients. Nup93 silencing inhibited BLCA cell proliferation, Wnt/β-catenin activation, and cancer cell stemness. Conversely, Nup93 overexpression promoted these effects. BML-284 partially rescued the reduction in cell growth and stemness markers caused by Nup93 knockdown. Nup93 knockdown also suppressed the tumor formation of BLCA cells in vivo. Nup93 regulates BLCA cell growth and stemness via the Wnt/β-catenin pathway, suggesting its potential as a prognostic marker and therapeutic target in BLCA.
Collapse
Affiliation(s)
- Zhe Wang
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Jing Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Lina Luo
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Chao Zhang
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Xiaomeng Huang
- Medical Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Shuo Liu
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Huaian Chen
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China
| | - Wenlong Miao
- Urology Department, The First Affiliated Hospital of Hebei North University, No. 12, Changqing Road, Zhangjiakou, 050051, Hebei Province, China.
| |
Collapse
|
7
|
Hasan E, Lewis CJ, Hernandez JG, Gentile P, Ferreira AM. Layer-by-layer assembly: advancing skin repair, one layer at a time. RSC Adv 2025; 15:13908-13923. [PMID: 40303355 PMCID: PMC12039284 DOI: 10.1039/d4ra08115c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Skin wound management remains a critical global healthcare challenge, with annual costs exceeding £30 billion. Traditional treatments like autografts face limitations in cost, availability, and recovery times. This review explores spray-assisted Layer-by-Layer (LbL) technology as a transformative approach for wound healing, emphasising its ability to deposit natural- and synthetic-polyelectrolytes such as chitosan, alginate, hyaluronic acid, and collagen into nanoscale coatings. These biocompatible multilayers integrate therapeutic agents to accelerate healing, reduce infections, and mimic native extracellular matrix structures. The work highlights emerging spray device innovations that optimise spray parameters to enhance cell viability, coverage, and clinical outcomes. While LbL techniques demonstrate versatility across substrates and scalability via immersion, spray, and microfluidic methods, challenges persist in manufacturing uniformity and clinical translation. The review underscores the urgent need for clinical trials to validate Lbl-based coatings in real-world settings and addresses gaps in portable, sustainable device development. By bridging advanced materials science with clinical practice, spray-assisted LbL technology offers a roadmap to overcome current wound care limitations, prioritising biocompatibility, cost-efficiency, and improved patient safety in regenerative medicine.
Collapse
Affiliation(s)
- Elias Hasan
- School of Engineering, Faculty of Science, Agriculture & Engineering, Newcastle University Newcastle upon Tyne UK
| | - Christopher J Lewis
- Northern Regional Burn Centre, Royal Victoria Infirmary Newcastle upon Tyne UK
| | | | - Piergiorgio Gentile
- School of Engineering, Faculty of Science, Agriculture & Engineering, Newcastle University Newcastle upon Tyne UK
| | - Ana M Ferreira
- School of Engineering, Faculty of Science, Agriculture & Engineering, Newcastle University Newcastle upon Tyne UK
| |
Collapse
|
8
|
Tsukamoto S. Natural products that target p53 for cancer therapy. J Nat Med 2025:10.1007/s11418-025-01906-6. [PMID: 40295432 DOI: 10.1007/s11418-025-01906-6] [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: 03/17/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Wild-type p53 acts as a tumor suppressor, but p53 is frequently mutated and inactivated in tumor cells, promoting cancer progression, invasion, and metastasis. Thus, compounds that reactivate p53 may be leveraged for cancer treatment, and the development of drugs targeting p53 reactivation is actively progressing. Notably, natural products exhibit diverse structures and biological activities and are used as therapeutic agents for various diseases worldwide. This review discusses the natural products that inhibit p53 degradation through p53-Mdm2 interaction, promote p53 reactivation by inducing conformational changes, and exhibit p53-dependent growth inhibition.
Collapse
Affiliation(s)
- Sachiko Tsukamoto
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Kumamoto, 862-0973, Japan.
| |
Collapse
|
9
|
Gu W. A bibliometric analysis of programmed cell death in oral cancer literature: research patterns and emerging trends (2000-2024). Discov Oncol 2025; 16:585. [PMID: 40261469 PMCID: PMC12014878 DOI: 10.1007/s12672-025-02410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Programmed cell death (PCD) plays a crucial role in oral cancer pathogenesis and treatment. However, a comprehensive bibliometric analysis of the global research landscape in this field has not been conducted. This study aims to analyze the evolution and current trends of PCD research in oral cancer from 2000 to 2024. METHODS Publications were retrieved from the Web of Science Core Collection database using relevant keywords related to oral cancer and PCD. VOSviewer 1.6.20 and CiteSpace 6.1R6 software were employed to conduct bibliometric analysis, including publication trends, citation analysis, co-authorship networks, keyword co-occurrence, and research hotspots. The time span was set from January 2000 to December 2024. RESULTS A total of 963 publications were identified and analyzed. The annual publication output showed a steady increase, with a significant growth rate after 2010, dividing the study period into three distinct phases. The most productive countries were China (58.42%), South Korea (12.27%), and Japan (10.04%), with China Medical University and Kaohsiung Medical University being the leading institutions. Research hotspots evolved from traditional apoptosis studies to emerging forms of PCD such as autophagy, ferroptosis, and pyroptosis. Keyword analysis revealed three major research clusters: basic molecular mechanisms (centered around ROS and oxidative stress), clinical aspects (including prognosis and cell proliferation), and cell death pathways. Citation burst analysis identified emerging trends in targeting multiple PCD pathways simultaneously for oral cancer therapy, with special focus on treatment resistance and survival. CONCLUSION This bibliometric analysis provides a comprehensive overview of global research trends in PCD and oral cancer over the past two decades. The findings highlight the shift from basic mechanistic studies focusing on apoptosis to more diverse PCD pathways and translational research. Emerging research directions include the exploration of synergistic mechanisms among multiple PCD pathways, development of AI-based personalized treatment plans, investigation of microenvironment regulation of PCD, and application of novel drug delivery systems. These trends demonstrate the field's evolution toward more integrated, personalized approaches in oral cancer treatment. This study offers valuable insights for researchers and funding agencies to identify research gaps and potential collaboration opportunities in this rapidly developing field.
Collapse
Affiliation(s)
- Wenli Gu
- Stomatological Hospital, School of Stomatology, Southern Medical University, S366 Jiangnan Boulevard, Haizhu District, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Babajnai A, Rahmani S, Asadi MJ, Gheytanchi E, Adibhesami G, Vakhshiteh F, Madjd Z. Molecular and phenotypic characterization of 5-FU resistant colorectal cancer cells: toward enrichment of cancer stem cells. Cancer Cell Int 2025; 25:154. [PMID: 40251609 PMCID: PMC12008981 DOI: 10.1186/s12935-025-03758-2] [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: 05/29/2024] [Accepted: 03/18/2025] [Indexed: 04/20/2025] Open
Abstract
Cancer stem cells (CSCs) as a subgroup of cells within a tumor capable of self-renewal, thereby driving tumor initiation and spread. Addressing treatment failures in cancer, linked to CSCs and their resistance mechanisms, requires effective preclinical models for testing targeted therapies. Caco2- and HT-29-resistant cells were generated by repeated treatment of cells with growing concentrations of 5-fluorouracil (5-FU) anticancer drug for an extended time. The sensitivity of 5-FU-resistant cells was evaluated by cytotoxicity assay. Stemness, epithelial-mesenchymal transition (EMT), migration and drug resistance characteristics were assessed through gene expression investigation by real-time PCR. The expression of CD44, CD133, and CD66 were evaluated by flow cytometry. To end, the bioinformatic analysis estimated the molecular function and biological pathways considering the differential expression of selected genes and proteins. 5-FU-exposed cells displayed increased resistance to 5-FU. The gene expression analysis showed an upregulation of stemness genes (KLF4, SOX2, OCT4, C-MYC), enhanced scavenging system, and elevated expression of CSC surface markers (CD44 and CD133) compared to parental cells. Additionally, pro-EMT genes (TWIST1, SNAIL1, ZEB1, Vimentin, and N-cadherin) were significantly upregulated compared to parental cells, with the downregulation of E-cadherin as an EMT suppressor gene reflected in increased migration capacity. Moreover, increased expression of ABC transporter genes (ABCB1, ABCC1) was observed, correlating with enhanced drug resistance. The bioinformatic analysis highlighted pathways related to microRNAs in cancer, cells pluripotency, and proteoglycans. Methods of drug exposure take priority over spheroid formation, particularly due to their enhanced efficacy in stemness, EMT, and surface markers. This positions them as a promising protocol for establishing experimental models of CSCs.
Collapse
Affiliation(s)
- Amirhesam Babajnai
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Rahmani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Jamal Asadi
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran
| | - Elmira Gheytanchi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Glavizh Adibhesami
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Liao L, Wang YX, Fan SS, Hu YY, Wang XC, Zhang X. The role and clinical significance of tumor-associated macrophages in the epithelial-mesenchymal transition of lung cancer. Front Oncol 2025; 15:1571583. [PMID: 40304000 PMCID: PMC12037373 DOI: 10.3389/fonc.2025.1571583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related mortality worldwide. Tumor-associated macrophages (TAMs) and epithelial-mesenchymal transition (EMT) are key drivers of lung cancer metastasis and drug resistance. M2-polarized TAMs dominate the immunosuppressive tumor microenvironment (TME) and promote EMT through cytokines such as TGF-β, IL-6, and CCL2. Conversely, EMT-transformed tumor cells reinforce TAM recruitment and M2 polarization through immunomodulatory factors such as CCL2 and ZEB1, thereby establishing a bidirectional interplay that fuels tumor progression. Current evidence on this interaction remains fragmented, and a comprehensive review of the TAM-EMT regulatory network and its therapeutic implications is lacking. This review systematically integrates the bidirectional regulatory mechanisms between TAMs and EMT, highlighting their roles in lung cancer progression. It also summarizes emerging therapeutic strategies targeting TAM polarization and the EMT process, emphasizing their potential for clinical translation. This study fills the gap in systematic reviews on the interaction between TAMs and EMT, providing a comprehensive theoretical foundation for future research and the development of novel lung cancer therapies.
Collapse
Affiliation(s)
- Lei Liao
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Ying-Xia Wang
- Department of Pathology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Su-Su Fan
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Ying-Yue Hu
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Xue-Chang Wang
- Department of Pharmacy, Anning First People’s Hospital, Anning, China
| | - Xuan Zhang
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
- Yunnan College of Modern Biomedical Industry, Kunming, China
| |
Collapse
|
12
|
Diehl B, Kirchhoff A, Hansmann F. CD86 is linked to apoptosis in canine histiocytic sarcoma. Front Vet Sci 2025; 12:1546047. [PMID: 40271486 PMCID: PMC12014657 DOI: 10.3389/fvets.2025.1546047] [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: 12/16/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
Immune checkpoints are critical for the regulation of tumor growth and regression. Recently an effect of CD80 and CD86 on tumor regression in canine cutaneous histiocytoma has been described. Further, the expression of MX dynamin like GTPase 1 (mx1) in cancer is linked to immune evasion. Thus, the present study aimed to investigate the effects of CD80 and CD86 in histiocytic sarcoma (HS), a rare and progressive malignancy in dogs and to elucidate the status of the interferon-I pathway. Twenty-two tissue samples of HS from skin, lung and liver of 15 dogs were used. Immunohistochemistry targeting CD80, CD86, programmed death-ligand 1 (PD-L1), survivin, cleaved caspase-3 (Casp-3), stimulator of interferon genes (STING) and mx1 was performed. Slides were digitized and analyzed with QuPath. The numbers of CD86- and Casp-3 expressing cells showed a positive correlation. In the skin and lung, numbers of CD80 immunolabeled cells were higher than for CD86, while CD80 and CD86 levels were comparable in the liver. In general, low numbers of PD-L1 immunolabeled tumor cells were detected. Intranuclear survivin expression was linked to Casp-3. Mx1 and STING were expressed in tumor cells. A possible link between CD86 and Casp-3 points to a role of CD86 in tumor cell death. The findings indicate relevant differences in CD80 and CD86 expression between organs and a function in histiocytic disease in dogs. Further, the expression of markers of the interferon-type-I pathway indicates a role in immune evasion.
Collapse
Affiliation(s)
- Benjamin Diehl
- Faculty of Veterinary Medicine, Institute of Veterinary Pathology, Leipzig University, Leipzig, Germany
| | | | - Florian Hansmann
- Faculty of Veterinary Medicine, Institute of Veterinary Pathology, Leipzig University, Leipzig, Germany
| |
Collapse
|
13
|
Bohec P, Dupuy F, Tishkova V, de Noray VS, Valignat MP, Theodoly O. Microvalve-based gradient generators to control flow-free, time zero and long-term conditions. LAB ON A CHIP 2025; 25:2043-2052. [PMID: 40070238 DOI: 10.1039/d4lc00901k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Experiments with gradients of soluble bioactive species have significantly advanced with microfluidic developments that enable cell observation and stringent control of environmental conditions. While some methodologies rely on flow to establish gradients, others opt for flow-free conditions, which is particularly beneficial for studying non-adherent and/or shear-sensitive cells. In flow-free devices, bioactive species diffuse either through resistive microchannels in microchannel-based devices, through a porous membrane in membrane-based devices, or through a hydrogel in gel-based devices. However, despite significant advancements over traditional methods such as Boyden chambers, these technologies have not been widely disseminated in biological laboratories, arguably due to entrenched practices and the intricate skills required for conducting microfluidic assays. Here, we developed microfluidic platforms integrating barriers with Quake-type pneumatic microvalves in place of microgrooves, membranes, or gels. One set of microvalves is used to maintain flow-free conditions and another set to regulate diffusion between a central channel housing the specimen of interest and sink/source reservoirs. This configuration enables stringent control over residual flows, precise spatial-temporal regulation of gradient formation, and exceptional gradient stability, maintained over extended periods via automated refilling of source and sink reservoirs. The gradient establishment was validated using fluorescent tracers with molar masses of 0.3-40 kDa, while cellular assays demonstrated the chemotactic response of primary human neutrophils swimming toward FMLP. The fabrication of microfluidic devices remains standardly demanding, but experimentation can be fully automated thanks to microvalves, making it accessible to non-expert users. This work presents a robust microfluidic approach for generating tunable gradients with stringent control over flow-free, time-zero, and long-term conditions and its automation and accessibility may promote adoption in academic and biomedical settings especially for non-adherent specimens.
Collapse
Affiliation(s)
- Pierre Bohec
- LAI, CNRS, INSERM, Turing Center for Living Systems, Aix Marseille Univ, Marseille, France
| | - Florian Dupuy
- LAI, CNRS, INSERM, Turing Center for Living Systems, Aix Marseille Univ, Marseille, France
| | | | | | - Marie-Pierre Valignat
- LAI, CNRS, INSERM, Turing Center for Living Systems, Aix Marseille Univ, Marseille, France
| | - Olivier Theodoly
- LAI, CNRS, INSERM, Turing Center for Living Systems, Aix Marseille Univ, Marseille, France
| |
Collapse
|
14
|
Mondal T, Chattopadhyay D, Saha Mondal P, Das S, Mondal A, Das A, Samanta S, Saha T. Fusobacterium nucleatum modulates the Wnt/β-catenin pathway in colorectal cancer development. Int J Biol Macromol 2025; 299:140196. [PMID: 39848378 DOI: 10.1016/j.ijbiomac.2025.140196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The Wnt/β-catenin signalling pathway normally maintains cellular and tissue homeostasis by regulating cellular differentiation and survival in a controlled manner. An aberrantly regulated Wnt/β-catenin signalling pathway can transform into an oncogenic pathway, which is associated with Colorectal cancer (CRC) as well as other cancers. CRC is one of the most frequently occurring gastrointestinal cancers worldwide. In CRC tissues, deregulation of Wnt/β-catenin pathway is observed, which indicates that this oncogenic pathway directly promotes CRC malignancy, cell migration, angiogenesis, chemoresistance, as well as shorter lifespan of a patient. Growing evidence suggests that human commensal microbes have a strong association with carcinogenesis, particularly the prevalence and high enrichment of Fusobacterium nucleatum in CRC progression. The Wnt/β-catenin pathway is one of the targeted pathways by F. nucleatum in CRC, where Fusobacterium adhesin attaches to E-cadherin to initiate infection. Also, Wnt/β-catenin pathway can be a potential target for the treatment of both CRC and F. nucleatum-positive CRC. Here, we discuss the underlying mechanisms of F. nucleatum-positive CRC development through modulation of Wnt/β-catenin signalling and its possibility for the application in targeted therapy of F. nucleatum-positive CRC.
Collapse
Affiliation(s)
- Tanushree Mondal
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Deepanjan Chattopadhyay
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Paromita Saha Mondal
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Sanjib Das
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Amalesh Mondal
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India; Department of Physiology, Katwa Collage, Katwa, Purba Bardhaman, West Bengal 713130, India
| | - Abhishek Das
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Subhasree Samanta
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Tanima Saha
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India.
| |
Collapse
|
15
|
Greene G, Zonfa I, Ravasz Regan E. A Boolean network model of hypoxia, mechanosensing and TGF-β signaling captures the role of phenotypic plasticity and mutations in tumor metastasis. PLoS Comput Biol 2025; 21:e1012735. [PMID: 40238833 PMCID: PMC12061430 DOI: 10.1371/journal.pcbi.1012735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 05/08/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
The tumor microenvironment aids cancer progression by promoting several cancer hallmarks, independent of cancer-related mutations. Biophysical properties of this environment, such as the stiffness of the matrix cells adhere to and local cell density, impact proliferation, apoptosis, and the epithelial to mesenchymal transition (EMT). The latter is a rate-limiting step for invasion and metastasis, enhanced in hypoxic tumor environments but hindered by soft matrices and/or high cell densities. As these influences are often studied in isolation, the crosstalk between hypoxia, biomechanical signals, and the classic EMT driver TGF-β is not well mapped, limiting our ability to predict and anticipate cancer cell behaviors in changing tumor environments. To address this, we built a Boolean regulatory network model that integrates hypoxic signaling with a mechanosensitive model of EMT, which includes the EMT-promoting crosstalk of mitogens and biomechanical signals, cell cycle control, and apoptosis. Our model reproduces the requirement of Hif-1α for proliferation, the anti-proliferative effects of strong Hif-1α stabilization during hypoxia, hypoxic protection from anoikis, and hypoxia-driven mechanosensitive EMT. We offer experimentally testable predictions about the effect of VHL loss on cancer hallmarks, with or without secondary oncogene activation. Taken together, our model serves as a predictive framework to synthesize the signaling responses associated with tumor progression and metastasis in healthy vs. mutant cells. Our single-cell model is a key step towards more extensive regulatory network models that cover damage-response and senescence, integrating most cell-autonomous cancer hallmarks into a single model that can, in turn, control the behavior of in silico cells within a tissue model of epithelial homeostasis and carcinoma.
Collapse
Affiliation(s)
- Grant Greene
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| | - Ian Zonfa
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| | - Erzsébet Ravasz Regan
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| |
Collapse
|
16
|
Zhou E, Yang JI, Habowski AN, Deschênes A, Belleau P, Ha T, Tzanavaris CJ, Boyd J, Hollweg CA, Zhu X, Tuveson DA, King DA. GATA6 Amplification Is Associated With Improved Survival in TP53-Mutated Unresectable Pancreatic Cancer. Pancreas 2025; 54:e303-e309. [PMID: 40262102 DOI: 10.1097/mpa.0000000000002431] [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: 05/23/2024] [Accepted: 11/01/2024] [Indexed: 04/24/2025]
Abstract
OBJECTIVES GATA6 expression is recognized as a favorable prognostic marker of pancreatic cancer, whereas TP53 is a poor prognostic marker. We evaluated treatment outcomes by genetic alterations in TP53 and GATA6 to determine the prognostic and predictive impact of co-alterations. MATERIALS AND METHODS A single institution retrospective analysis was performed on patients diagnosed with pancreatic ductal adenocarcinoma between 2014 and 2023. TP53 genotype and GATA6 amplification status were included in an analysis of overall survival (OS) and progression-free survival (PFS). Previously published patient-derived organoids were used to investigate correlation between genetic status and drug sensitivity. RESULTS Patients with TP53 mutations had worse OS compared with the wild-type TP53 population. Patients with GATA6 amplification had better OS and a trend toward better PFS than the nonamplified population. Among patients with a TP53 mutation, patients with GATA6 co-alteration had longer OS compared with those who were not GATA6 amplified. In contrast, among patients who were TP53 wild-type, the presence or absence of a GATA6 amplification did not impact OS or PFS. GATA6 genotype was not associated chemotherapy drug response in an organoid pharmacotyping model. CONCLUSIONS We found that GATA6 amplification appeared to attenuate poor prognosis observed in TP53-mutant patients regardless of the type of standard chemotherapy received, suggesting the GATA6 amplification is a prognostic biomarker but not a predictive biomarker of standard-of-care chemotherapy.
Collapse
Affiliation(s)
- Edward Zhou
- Northwell Health Cancer Institute, New York, NY
| | | | | | | | - Pascal Belleau
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Taehoon Ha
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Chris J Tzanavaris
- Division of Pulmonary, Critical Care, and Sleep Medicine, Northwell Health, New York, NY
| | - Jeff Boyd
- Northwell Health Cancer Institute, New York, NY
| | - Christopher A Hollweg
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, NY
| | - Xinhua Zhu
- Northwell Health Cancer Institute, New York, NY
| | | | | |
Collapse
|
17
|
Sanjida S, Garvey G, Bainbridge R, Diaz A, Barzi F, Holzapfel S, Chen MY, Collin H, Fatima Y, Hou XY, Ward J. Prevalence of surgery in Indigenous people with cancer: a systematic review and meta-analysis. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2025; 57:101527. [PMID: 40225852 PMCID: PMC11992426 DOI: 10.1016/j.lanwpc.2025.101527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/02/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025]
Abstract
Background As cancer incidence increases globally, so does the prevalence of cancer among Indigenous peoples. Indigenous peoples face significant barriers to healthcare, including access to and uptake of surgery. To date, the synthesis of access to and uptake of surgery for Indigenous peoples living with cancer has not yet been reported. Methods We conducted a systematic literature review and meta-analysis of access to and uptake of surgery for Indigenous peoples in Canada, Australia, New Zealand, and the United States. Five databases were searched to identify studies of Indigenous adults with cancer and those who received surgery. The Joanna Briggs Institute critical appraisal tools were used to assess the quality and inclusion of articles. Random effect meta-analyses were conducted to estimate the pooled prevalence of surgery in Indigenous people with cancer. Findings Of the 52 studies in the systematic review, 38 were included in the meta-analysis. The pooled prevalence of surgery in Indigenous people with cancer was 56.2% (95% confidence interval (CI): 45.4-66.7%), including 42.8% (95% CI: 36.3-49.5%) in the Native Hawaiian population, 44.5% (95% CI: 38.7-50.3%) in the Inuit and 51.5% (95%CI: 36.8-65.9%) in Aboriginal and Torres Strait Islander people. Overall, Indigenous people received marginally less cancer surgery than non-Indigenous people (3%, 95% CI: 0-6%). Indigenous people were 15% (95% CI: 6-23%) less likely to receive surgery than non-Indigenous people for respiratory cancers. Remoteness, travel distance, financial barriers, and long waiting times to receive surgery were factors cited as contributing to lower access to surgery for Indigenous people compared to non-Indigenous people. Interpretation Efforts to improve access and use of cancer services and surgery for Indigenous peoples should be multilevel to address individual factors, health services and systems, and structural barriers. These determinants need to be addressed to expedite optimal care for Indigenous peoples, especially those living in outer metropolitan areas. Funding The Research Alliance for Urban Goori Health (RAUGH) funded this project. GG was funded by an NHMRC Investigator Grant (#1176651).
Collapse
Affiliation(s)
- Saira Sanjida
- Poche Centre for Indigenous Health, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Queensland, Australia
| | - Gail Garvey
- School of Public Health, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Queensland, Australia
| | - Roxanne Bainbridge
- Indigenous Future Centre, Faculty of Business, Economics and Law, The University of Queensland, Queensland, Australia
| | - Abbey Diaz
- School of Public Health, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Queensland, Australia
- Yardhura Walani National Centre for Aboriginal and Torres Strait Islander Wellbeing Research, The Australian National University, Australian Capital Territory, Australia
| | - Federica Barzi
- Poche Centre for Indigenous Health, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Queensland, Australia
| | - Sherry Holzapfel
- Aboriginal and Torres Strait Islander Health, Metro North Hospital and Health Service, Queensland, Australia
| | - Michael Y. Chen
- School of Medicine, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Queensland, Australia
| | - Harry Collin
- Royal Brisbane and Women’s Hospital, Queensland, Australia
| | - Yaqoot Fatima
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Xiang-Yu Hou
- Broken Hill University Department of Rural Health, The University of Sydney, New South Wales, Australia
| | - James Ward
- Poche Centre for Indigenous Health, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Queensland, Australia
| |
Collapse
|
18
|
Avila E, Hernández-Monterde LD, Cedro-Tanda A, Lizardi-Aguilera TM, Barrera D, Villegas-Rodriguez FV, García-Quiroz J, Díaz L, Larrea F. Transcriptomic profile induced by calcitriol in CaSki human cervical cancer cell line. PLoS One 2025; 20:e0319812. [PMID: 40168262 PMCID: PMC11960991 DOI: 10.1371/journal.pone.0319812] [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: 05/17/2024] [Accepted: 02/09/2025] [Indexed: 04/03/2025] Open
Abstract
The vitamin D endocrine system, primarily mediated by its main metabolite calcitriol and the vitamin D receptor (VDR), plays a critical role in numerous human physiological processes, ranging from calcium metabolism to the prevention of various tumors, including cervical cancer. In this study, we comprehensively investigated the genomic regulatory effects of calcitriol in a cervical cancer model. We examined the transcriptional changes induced by calcitriol in CaSki cells, a cervical cell line harboring multiple copies of HPV16, the primary causal agent of cervical cancer. Our microarray findings, revealed that calcitriol regulated over 1000 protein-coding genes, exhibiting a predominantly repressive effect on the CaSki cell transcriptome by suppressing twice as many genes as it induced. Calcitriol decreased EPHA2 and RARA expression while inducing KLK6 and CYP4F3 expression in CaSki cells, as validated by qPCR and Western blot. Functional analysis demonstrated that calcitriol effectively inhibited key processes involved in cancer progression, including cell proliferation and migration. This was further supported by the significant downregulation of MMP7 and MMP13 mRNA levels. Our microarray results also showed that, in addition to its effects on protein-coding genes, calcitriol significantly regulates non-coding RNAs, altering the expression of approximately 400 non-coding RNAs, including 111 microRNA precursors and 29 mature microRNAs, of which 17 were upregulated and 12 downregulated. Notably, among these calcitriol-regulated microRNAs are some involved in cervical cancer biology, such as miR-6129, miR-382, miR-655, miR-211, miR-590, miR-130a, miR-301a, and miR-1252. Collectively, these findings suggest that calcitriol exhibits a significant antitumor effect in this advanced cervical cancer model by blocking critical processes for tumor progression, underscoring the importance of maintaining adequate vitamin D nutritional status.
Collapse
Affiliation(s)
- Euclides Avila
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis David Hernández-Monterde
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Tomas Misael Lizardi-Aguilera
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - David Barrera
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Francisco Vladimir Villegas-Rodriguez
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Fernando Larrea
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
19
|
Chou MY, Lin-Shiau SY. Non-steroidal anti-inflammatory drugs (NSAIDs) regimens enhance synergistic selective anticancer efficacy of chemotherapeutic agents on cultured cells. J Dent Sci 2025; 20:1175-1195. [PMID: 40224060 PMCID: PMC11993116 DOI: 10.1016/j.jds.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/19/2025] [Indexed: 04/15/2025] Open
Abstract
Background/purpose Cancer incidences are rising, presenting challenges due to severe side effects and resistance of chemotherapeutic agents. To address these issues, we propose a strategy involving pharmaceutical compositions of PTM (Phytopolyphenols, Targeting drugs, Metals) regimens. This study aimed to investigate NSAIDs (Non-steroidal anti-inflammatory drugs)-PTM regimens enhancing anticancer selectivity and efficacy of chemotherapeutic agents on cultured cancer cells. Materials and methods Effects of drugs on proliferation of cultured cancer cells and pathogens was assessed using MTT assay and optical density at 600 nm (OD600) respectively. Synergistic effects of drug combinations were determined using combination index and efficacy index. ATPase activity was assayed using a colorimetric method. Results NSAIDs-PTM regimens demonstrated selective and synergistic anticancer effects. They also enhanced anticancer selectivity and efficacy of Cisplatin, 5-Fluorouracil, and Methotrexate. The most effective NSAIDs-PTM regimens increased anticancer efficacy by 16, 4, and 23 fold against oral, lung, and colon cancer cell lines, respectively. Additionally, these NSAIDs-PTM regimens enhanced selective anticancer efficacy of Cisplatin, 5-Fluorouracil, and Methotrexate by 8-21 fold on the three cancer cells. Furthermore, all regimens exhibited synergistic anti-efflux pump ATPase activity and antibacterial effects against four cultured pathogens. Conclusion The findings indicate that NSAIDs-PTM regimens not only possess synergistic and selective anticancer and antibacterial properties but also enhance anticancer selectivity and efficacy of Cisplatin, 5-Fluorouracil, and Methotrexate. Notably, all regimens exhibited anti-efflux pump ATPase, which may help overcome multidrug resistance in cancer treatment. Given that all components of PTM regimens are clinically effective and safe, further clinical studies are warranted.
Collapse
Affiliation(s)
- Ming-Yung Chou
- School of Dentistry, College of Oral Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shoei-Yn Lin-Shiau
- School of Dentistry, College of Oral Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
20
|
Agrawal R, Sengupta S. p53 regulates DREAM complex-mediated repression in a p21-independent manner. EMBO J 2025; 44:2279-2297. [PMID: 40038454 PMCID: PMC12000331 DOI: 10.1038/s44318-025-00402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 01/18/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The DREAM repressor complex regulates genes involved in the cell cycle and DNA repair, vital for maintaining genome stability. Although it mediates p53-driven repression through the canonical p53-p21-Rb axis, the potential for p53 to directly regulate DREAM targets independently of its transcriptional activity has not been explored. Here, we demonstrate that in asynchronously growing cells, p53 loss leads to greater de-repression of DREAM targets compared to p21 loss alone. Both wild-type and transactivation-deficient p53 mutants are capable of repressing DREAM targets, suggesting a transactivation-independent "non-canonical" repression mechanism. These p53 variants bind p130/p107, irrespective of their phosphorylation status, while cancer-associated p53 mutants disrupt DREAM complex function by sequestering E2F4. Re-ChIP analysis shows co-recruitment of p53 and E2F4 to known and newly identified DREAM target promoters, indicating direct repression of these targets by p53. These findings reveal a novel, transactivation-independent mechanism of p53-mediated repression, expanding our understanding of p53's tumor-suppressive functions and suggesting DREAM complex targeting as potential future avenues in cancer therapy.
Collapse
Affiliation(s)
- Ritu Agrawal
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sagar Sengupta
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India.
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), PO: NSS, Kalyani, 741251, India.
| |
Collapse
|
21
|
Romero-López MJ, Jiménez-Wences H, Nuñez-Martínez HN, Cruz-De la Rosa MI, Alarcón-Millán J, Fernández-Tilapa G. Overexpression of miR-23b-3p+miR-218-5p+miR-124-3p differentially modifies the transcriptome of C-33A and CaSki cells and the regulation of cellular processes involved in the progression of cervical cancer. Comput Biol Med 2025; 188:109886. [PMID: 40010175 DOI: 10.1016/j.compbiomed.2025.109886] [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: 09/30/2024] [Revised: 01/12/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Dysregulation of tumor suppressor miRNAs (tsmiRs) is associated with tumor progression in cancer. miR-23b-3p, miR-218-5p and miR-124-3p are tsmiRs in cervical cancer (CC) and regulate the translation of genes involved in metastasis-related biological processes. OBJECTIVE To analyze transcriptome changes in cervical cancer cell lines (C-33A HPV-negative and CaSki HPV-positive) overexpressing miR-23b-3p + miR-218-5p + miR-124-3p, to identify specific target transcripts common to all three miRNAs, as well as signaling pathways and cellular processes related to tumor progression. METHODS The transcriptome of C-33A and CaSki cells transfected with miR-23b-3p + miR-218-5p + miR-124-3p was analyzed by RNA-seq. Differentially expressed genes (DEGs) were subjected to Gene Ontology analysis on the DAVID platform. The function of under-regulated genes was analyzed on the GEPIA 2.0, Kaplan-Meier plotter and STRING platforms. On the TargetScanHuman platform it was determined which transcripts have MREs for miR-23b-3p, miR-218-5p and/or miR-124-3p in their 3'UTR region. RESULTS Simultaneous overexpression of miR-218-5p, miR-124-3p and miR-23b-3p induced changes in global gene expression in C-33A and CaSki cells. In C-33A cells, DEGs included 45 over- and 172 under-regulated transcripts; in CaSki, 125 transcripts were over- and 84 under-regulated. The under-regulated transcripts enrich proliferation, migration, apoptosis and angiogenesis; 20 of these genes are associated with overall survival (OS) in women with CC, and 18 of the 20 mRNAs have MREs for one, two or all three miRNAs. CONCLUSIONS miR-23b-3p + miR-218-5p + miR-124-3p, differentially modify global gene expression in C-33A and CaSki cells. The results indicate that these miRNAs act synergistically and modulate CC progression through individual and shared targets by two or all three miRNAs.
Collapse
Affiliation(s)
- Manuel Joaquín Romero-López
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México
| | - Hilda Jiménez-Wences
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México; Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México
| | - Hober Nelson Nuñez-Martínez
- Instituto de Fisiología Celular, Departamento de Genética Molecular, Universidad Nacional Autónoma de México, Ciudad de México, C.P. 04510, Mexico
| | - Merlin Itsel Cruz-De la Rosa
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México
| | - Judit Alarcón-Millán
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México; Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México
| | - Gloria Fernández-Tilapa
- Laboratorio de Investigación Clínica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México; Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo, Guerrero. C.P. 39090. México.
| |
Collapse
|
22
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [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: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
23
|
Yan X, Xu L, Qi C, Chang Y, Zhang J, Li N, Shi B, Guan B, Hu S, Huang C, Wang H, Chen Y, Xu X, Lu J, Xu G, Chen C, Li S, Chen Y. Brazilin alleviates acute lung injury via inhibition of ferroptosis through the SIRT3/GPX4 pathway. Apoptosis 2025; 30:768-783. [PMID: 39720978 DOI: 10.1007/s10495-024-02058-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 12/26/2024]
Abstract
Ferroptosis is a novel type of programmed cell death dependent on iron and is characterized by the accumulation of lipid peroxides, which is involved in acute lung injury (ALI). Brazilin, an organic compound known for its potent antioxidant and anti-inflammatory properties, has not been thoroughly studied for its potential impact on lipopolysaccharide (LPS)-induced ALI. Here, we found that pretreatment of brazilin mitigated LPS-induced lung injury and inflammation by inhibiting mitochondrial oxidative stress and ferroptosis, both in vivo and in vitro. Sirtuin 3 (SIRT3) was identified as a downstream target of brazilin, and overexpression of SIRT3 mirrored the protective effects of brazilin against LPS-induced ALI. Additionally, SIRT3 contributed to the upregulation, mitochondrial translocation and deacetylation of glutathione peroxidase 4 (GPX4). Through screening potential acetylation sites on GPX4, we identified lysine 148 (K148) as the residue deacetylated by SIRT3. Mutating the acetylation site of GPX4 within mitochondria (mitoGPX4-K148R) reduced LPS or SIRT3 knockdown-induced GPX4 acetylation, oxidative stress, and ferroptosis, ultimately ameliorating ALI. In conclusion, our study demonstrates the beneficial effects of brazilin in treating LPS-induced ALI. Brazilin enhances SIRT3 expression, which in turn deacetylates and facilitates the mitochondrial translocation of GPX4, thereby reducing mitochondrial oxidative stress and ferroptosis. These findings suggest that the SIRT3/GPX4 pathway may represent a critical mechanism, and brazilin emerges as a promising therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Xiaopei Yan
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Li Xu
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Chang Qi
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiling Chang
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Jiangsu, China
| | - Juanjuan Zhang
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ning Li
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Baoyu Shi
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Bo Guan
- Department of Geriatrics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Siming Hu
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Chao Huang
- Ministry of Science and Technology, Public Experimental Department, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Hui Wang
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ying Chen
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xiao Xu
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jian Lu
- Department of Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Guopeng Xu
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Chao Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu, China.
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Yuqiong Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
24
|
Kim M, Lee M, Lee A, Choi BO, Park WC, Kim SH, Lee J, Kang J. Correlating p53 immunostaining patterns with somatic TP53 mutation and functional properties of mutant p53 in triple-negative breast cancer. Histopathology 2025. [PMID: 40162573 DOI: 10.1111/his.15453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/21/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025]
Abstract
AIMS Immunohistochemical (IHC) staining of p53 is a potential marker for TP53 mutations in various cancers. However, criteria for predicting TP53 mutations in triple-negative breast cancer (TNBC) using p53 IHC staining are not yet established. We aim to correlate p53 IHC expression patterns with TP53 mutation status in TNBC. METHODS AND RESULTS A total of 113 TNBC cases were analysed for p53 IHC staining pattern and somatic TP53 mutation using whole-exome sequencing. Functional properties of TP53 mutations were determined using the National Cancer Institute (NCI) TP53 database. P53 IHC patterns were categorized as nuclear overexpression (n = 58), null pattern (n = 40), wildtype (n = 15), cytoplasmic (n = 5), and subclonal (n = 5). The cutoff for predictive p53 nuclear overexpression was determined to be 80%, which strongly correlated with TP53 mutations. Notably, p53 overexpression had a positive predictive value (PPV) of 83% for missense or in-frame mutations, while the null pattern showed a PPV of 85% for detecting nonsense, frameshift, or splicing mutations. P53 overexpression was significantly linked to missense mutations within the DNA-binding domain (DBD) exhibiting gain-of-function (GOF) or dominant-negative effect (DNE). Cases exhibiting cytoplasmic expression correlated with nonsense or frameshift mutations in the DBD, nuclear localization signal (NLS), or splice sites. Cases with subclonal p53 staining patterns were associated with TP53 mutations. CONCLUSION Our study proposes newly defined criteria for interpreting p53 immunostaining patterns in TNBC, potentially allowing for the prediction of TP53 mutation types and their functional implications.
Collapse
Affiliation(s)
- Meejeong Kim
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Miseon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Byung-Ock Choi
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woo-Chan Park
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Hun Kim
- Department of Radiology, Seoul Saint Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jun Kang
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
25
|
Wu H, Shang J, Bao Y, Liu H, Zhang H, Xiao Y, Li Y, Huang Z, Cheng X, Ma Z, Zhang W, Mo P, Wang D, Zhang M, Zhan Y. Identification of a novel prognostic marker ADGRG6 in pancreatic adenocarcinoma: multi-omics analysis and experimental validation. Front Immunol 2025; 16:1530789. [PMID: 40226617 PMCID: PMC11986822 DOI: 10.3389/fimmu.2025.1530789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Background Pancreatic adenocarcinoma (PAAD) ranks among the most lethal malignancies worldwide. Current treatment options have limited efficacy, underscoring the need for new therapeutic targets. Methods This study employed a multi-omics analytical framework to delve into the expression profiles and prognostic implications of ADGRG6 within the pan-cancer dataset of The Cancer Genome Atlas (TCGA) database, highlighting the prognostic value and potential carcinogenic role of ADGRG6 in PAAD, which was further validated using data from multiple PAAD cohorts in the Gene Expression Omnibus (GEO) database. To assess the role of ADGRG6 in the tumor microenvironment of PAAD, we evaluated immune infiltration using CIBERSORT, ssGSEA, xCell and Tracking Tumor Immunophenotype (TIP), and utilized single-cell sequencing data to explore cell-cell interactions. Further cellular and animal experiments, such as colony formation assay, transwell assay, western blot, real-time PCR, and tumor xenograft experiments, were used to investigate the effect of ADGRG6 on the proliferation, metastatic potential and immune marker expression of PAAD and the underlying mechanisms. Results ADGRG6 emerged as a potential prognostic biomarker and a therapeutic target for PAAD, which was further corroborated by data extracted from multiple PAAD cohorts archived in the GEO database. Single-cell sequencing and immune infiltration analyses predicted the positive correlation of ADGRG6 with the infiltration of immune cells and with the interaction between malignant cells and fibroblasts/macrophages within the PAAD microenvironment. In vitro cell assays demonstrated that ADGRG6 promoted the proliferation, metastatic potential and immune marker expression of PAAD cells by increasing protein level of mutated p53 (mutp53), which activated a spectrum of gain-of-functions to promote cancer progression via the EGFR, AMPK and NF-κB signaling cascades. Furthermore, subcutaneous xenograft experiments in mice demonstrated that ADGRG6 knockdown substantially suppressed the growth of engrafted PAAD tumors. Conclusions ADGRG6 may serve as a novel prognostic marker and a therapeutic targets for PAAD, playing a crucial role in the proliferation, metastasis, and immune marker regulation of PAAD through elevating protein level of mutated p53.
Collapse
Affiliation(s)
- Han Wu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Jin Shang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yuanyan Bao
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Huajie Liu
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Haoran Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yaosheng Xiao
- Department of Infectious Disease, Xiang’an Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Yangtaobo Li
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Zhaozhang Huang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaolei Cheng
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zixuan Ma
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenqing Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Daxuan Wang
- Provincial College of Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Mingqing Zhang
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Yanyan Zhan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
26
|
Gu P, Chen J, Xin J, Chen H, Zhang R, Chen D, Zhang Y, Shao S. Network pharmacology-based investigation of the pharmacological mechanisms of diosgenin in nonalcoholic steatohepatitis. Sci Rep 2025; 15:10351. [PMID: 40133701 PMCID: PMC11937522 DOI: 10.1038/s41598-025-95154-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
The prevalence of nonalcoholic steatohepatitis (NASH) is rising annually, posing health and economic challenges, with limited treatments available. Diosgenin, a natural steroidal compound found in various plants, holds potential as a therapeutic candidate. Recent studies have confirmed diosgenin's anti-inflammatory and metabolism-modulating properties. However, its therapeutic effects on NASH and the underlying mechanisms are still unclear. This study aims to explore diosgenin's protective effects and pharmacological mechanisms against NASH using network pharmacology, molecular docking, and experimental validation. We gathered potential targets of diosgenin and NASH from various databases to generate protein-protein interaction (PPI) networks. GO and KEGG pathway enrichment analyses identified key targets and mechanisms. Molecular docking confirmed the binding capacity between diosgenin and core target proteins. Additionally, a NASH cell model was developed to validate the pharmacological effects of diosgenin. Our investigation identified nine key targets (ALB, AKT1, TP53, VEGFA, MAPK3, EGFR, STAT3, CASP3, IGF1) that interact with diosgenin. Molecular docking indicated potential bindings interactions, while enrichment analyses revealed that diosgenin may enhance fatty acid metabolism via the PI3K-Akt pathway. Cellular experiments confirmed that diosgenin activates this pathway, reduces SCD1 expression, and decreases triglyceride and IL-6 levels. Our study elucidates that diosgenin may ameliorate triglyceride deposition and inflammation through the PI3K-Akt pathway.
Collapse
Affiliation(s)
- Peiyuan Gu
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
| | - Juan Chen
- Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jingxin Xin
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Huiqi Chen
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
| | - Ran Zhang
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
| | - Dan Chen
- Department of Electrocardiographic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yuhan Zhang
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China.
| | - Shanshan Shao
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China.
| |
Collapse
|
27
|
Jasim SA, Pallathadka H, Sivaprasad GV, Kumar A, Mustafa YF, Mohammed JS, Eldesoqui M, Pramanik A, Abdukarimovna RK, Zwamel AH. New approaches of chimeric antigen receptor (CAR)-immune cell-based therapy in gastric cancer; highlight CAR-T and CAR-NK. Funct Integr Genomics 2025; 25:72. [PMID: 40133688 DOI: 10.1007/s10142-025-01584-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/14/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
One characteristic that makes gastric cancer (GC) against other cancers is the intricate immune system's reaction, particularly to tenacious inflammation. Consequently, the immunological function is essential to the growth of this malignancy. Tumor immunotherapy has yielded several encouraging outcomes, but despite this, different patients continue to not respond to treatment, and a far larger number become resistant to it. Also, activated CAR-T cells express a majority of immunological checkpoint factors, containing PD1, CTLA4, and LAG3, which counteracts the anti-tumor actions of CAR-T cells. Moreover, cytokine release syndrome is one of the possible adverse responses of CAR-T cell therapy. Therefore, producing universal allogeneic T lymphocytes with potent anti-tumor activity is essential. This study demonstrates current research on this cutting-edge technology, including the composition and mode of action of CAR-NK and CAR-T cells in GC. Also, in this study, we examined recent studies about various specific GC biomarkers that target CAR-T cells and CAR-NK cells.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | | | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Ashwani Kumar
- Department of Life Scienzces, School of Sciences, Jain (Deemed-to-Be) University, Bengaluru, Karnataka, 560069, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul-41001, Iraq
| | | | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, Almaarefa University, 13713, DiriyahRiyadh, Saudi Arabia.
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Divison of Research and Innovation, Uttaranchal University Dehradun, Dehradun, Uttarakhand, India
| | - Rakhimova Khusnidakhon Abdukarimovna
- Department of Folk Medicine and Pharmacology, Fergana Public Health Medical Institute, Fergana, Uzbekistan
- Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
28
|
Alamillo-Vásquez JA, Pérez-Torres CA, Ibarra-Laclette E, Ramón-Farías F, Nicasio-Torres P, Alatorre-Cobos F. Secondary Metabolites from Croton Species and Their Biological Activity on Cell Cycle Regulators. Metabolites 2025; 15:216. [PMID: 40278345 PMCID: PMC12029420 DOI: 10.3390/metabo15040216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Plant-based traditional medicine integrates beliefs, knowledge, and practices to prevent and treat multiple diseases. Croton is a large and worldwide-spread genus belonging to Euphorbiaceae, a family well known for comprising many species with medicinal properties due to its high diversity of phytochemical constituents with biological activities. Among the various benefits of Croton species in traditional medicine, its use in cancer treatment has recently received significant attention from the scientific community. This review provides a general overview of different studies on the Croton genus in the research for alternative cancer treatments and the impact of its secondary metabolite catalog on cell cycle targets. Our analysis indicates that just under 30 secondary metabolites have been identified so far in latex and extracts obtained from leaves, twigs, or bark from 22 different Croton species. Based on standard assays using cell lines or human platelets, these molecules show multiple biological activities mainly compromising cell viability and cell cycle progression, supporting the ethnobotanical use of Croton species for cancer treatment. Several studies indicate that Croton metabolites target CDK-cyclin complexes and signaling routes that trigger apoptosis; however, further studies are needed to better understand the molecular mechanisms underlying Croton metabolites' effects and their accurate future applications in cancer treatment.
Collapse
Affiliation(s)
- Jorge Augusto Alamillo-Vásquez
- Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico; (J.A.A.-V.); (E.I.-L.)
| | - Claudia-Anahí Pérez-Torres
- SECIHTI-Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico
| | - Enrique Ibarra-Laclette
- Red de Estudios Moleculares Avanzados (REMAV), Instituto de Ecología, A.C. (INECOL), Carretera Antigua a Coatepec 351, Col. El Haya, Xalapa 91073, Veracruz, Mexico; (J.A.A.-V.); (E.I.-L.)
| | - Feliza Ramón-Farías
- Facultad de Ciencias Biológicas y Agropecuarias, Universidad Veracruzana, Córdoba 94500, Veracruz, Mexico;
| | - Pilar Nicasio-Torres
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62790, Morelos, Mexico;
| | - Fulgencio Alatorre-Cobos
- SECIHTI-Unidad de Biología Integrativa Centro de Investigación Científica de Yucatán, A.C., Calle 43 No. 130 × 32 y 34, Col. Chuburná de Hidalgo, Mérida 97205, Yucatán, Mexico
| |
Collapse
|
29
|
Gama JM, Oliveira RC. CD44 and Its Role in Solid Cancers - A Review: From Tumor Progression to Prognosis and Targeted Therapy. FRONT BIOSCI-LANDMRK 2025; 30:24821. [PMID: 40152366 DOI: 10.31083/fbl24821] [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: 05/13/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 03/29/2025]
Abstract
Cluster of differentiation 44 (CD44) is a transmembrane protein expressed in normal cells but overexpressed in several types of cancer. CD44 plays a major role in tumor progression, both locally and systemically, by direct interaction with the extracellular matrix, inducing tissue remodeling, activation of different cellular pathways, such as Akt or mechanistic target of rapamycin (mTOR), and stimulation of angiogenesis. As a prognostic marker, CD44 has been identified as a major player in cancer stem cells (CSCs). CSCs with a CD44 phenotype are associated with chemoresistance, alone or in combination with other CSC markers, such as CD24 or aldehyde dehydrogenase 1 (ALDH1), and may be used for patient stratification. In the therapy setting, CD44 has been explored as a viable target, directly or indirectly. It has revealed promising potential, paving the way for its future use in the clinical setting. Immunohistochemistry effectively detects CD44 overexpression, enabling patients to be accurately selected for surgery and targeted anti-CD44 therapies. In this review, we highlight the properties of CD44, its expression in normal and tumoral tissues through immunohistochemistry and potential treatment options. We also discuss the clinical significance of this marker and its added value in therapeutic decision-making.
Collapse
Affiliation(s)
- João Martins Gama
- Serviço de Anatomia Patológica, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-313 Porto, Portugal
| | - Rui Caetano Oliveira
- Centro de Investigação em Meio Ambiente, Genética e Oncobiologia-CIMAGO, Faculdade de Medicina, Universidade de Coimbra, 3004-535 Coimbra, Portugal
- Centro de Anatomia Patológica Germano de Sousa, 3000-377 Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
30
|
Dong Y, Sheng G, Chen W. TPX2 knockdown mediates p53 activation to induce autophagy and apoptosis for anti-colorectal cancer effects. J Recept Signal Transduct Res 2025:1-13. [PMID: 40116489 DOI: 10.1080/10799893.2025.2470180] [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: 07/22/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 03/23/2025]
Abstract
Colorectal cancer (CRC) exhibits high morbidity and mortality worldwide. Targeting protein for Xenopus kinesin-like protein 2 (TPX2) impacts various cancers; however, mechanism of TPX2 in CRC remains unclear. Xenograft nude mouse models were constructed by subcutaneous injection of HCT116 cells with sh-NC, sh-TPX2, OE-NC, and OE-TPX2 transfection. Following the test of tumor growth, immunohistochemistry and TUNEL staining were done. In vitro, HCT116, RKO, and SW480 cells were divided into sh-NC, sh-TPX2, and sh-TPX2 + 3-methyladenine (3-MA, autophagy inhibitor) groups. Further, sh-p53 and rapamycin (RA, autophagy agonist) were added in HCT116 cells. EdU staining, flow cytometry, transparent electron microscopy, and Western blot were performed. Comparing with sh-NC group, sh-TPX2 inhibited tumor growth and Ki67 expression, and increased LC3-II expression and apoptosis, whereas OE-TPX2 group presented an opposite trend. In vitro, HCT116 and RKO cells in sh-TPX2 group enhanced apoptosis and LC3 II/LC3 I expression, and inhibited proliferation and P62 expression, which were reversed after further 3-MA intervention. The above results were not found in SW480 cells. Moreover, compared to sh-TPX2 group, sh-TPX2 + RA group enhanced apoptosis and autophagy, and suppressed the proliferation of HCT116 cells, which were reversed following further sh-p53 intervention. Therefore, sh-TPX2 mediated p53 activation to induce autophagy for anti-CRC effects, providing new ideas for CRC treatment.
Collapse
Affiliation(s)
- Yunfei Dong
- Department of Anorectal, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Guixian Sheng
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
31
|
de Albuquerque RDDG, León-Vargas FR, Carrasco-Montañez DD, Carranza-Gonzales JL, Ramos-Rivas YVS. A Review on Phytochemistry and Recent Pharmacology of Dragon's Blood (Croton lechleri), a Multifunctional Ethnomedicinal Resource from the Amazon Forest. PLANTA MEDICA 2025. [PMID: 40043740 DOI: 10.1055/a-2551-5681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Croton lechleri, commonly known as "Sangre de Drago", is a widely utilized ethnomedicinal resource in the Amazon region, known for its diverse bioactive properties. These include wound-healing activity, anti-inflammatory effects, antitumor activity, and other therapeutic benefits. Despite its extensive traditional use, a comprehensive review of the scientific studies conducted over the past two decades is lacking, which hinders a thorough understanding of the chemical and pharmacological characteristics of this species. Hence, this review is essential to inform researchers and readers about the current state of knowledge in this field. A systematic search was conducted using databases such as Scopus and Google Scholar, yielding 33 relevant articles focusing on the phytochemistry and recent pharmacological investigations of C. lechleri. These studies identify proanthocyanidins as the predominant phytochemical group in terms of relative quantity. Additionally, other significant phytochemical groups include alkaloids, diterpenoids, phytosteroids, saponins, phenolics, and polyphenolics. The pharmacological studies reviewed highlight several potential therapeutic effects of C. lechleri, particularly those associated with its resin. These effects include wound-healing, antitumor, anti-inflammatory, and gastrointestinal benefits, among others. The findings underscore the remarkable medicinal importance of this species, supporting its continued investigation and potential therapeutic applications.
Collapse
Affiliation(s)
- Ricardo D D G de Albuquerque
- Laboratório de Tecnologia em Produtos Naturais, Universidade Federal Fluminense, Santa Rosa, Niterói, RJ, Brazil
| | - Frank R León-Vargas
- Departamento de Ingeniería Química, Facultad de Ingeniería Química, Universidad Nacional de la Amazonia Peruana, Iquitos, Loreto, Peru
| | - Daniel D Carrasco-Montañez
- Departamento de Ingeniería Química, Facultad de Ingeniería Química, Universidad Nacional de la Amazonia Peruana, Iquitos, Loreto, Peru
| | - Jorge L Carranza-Gonzales
- Departamento de Ingeniería de Alimentos, Facultad de Industrias Alimentarias, Universidad Nacional de la Amazonia Peruana, Iquitos, Peru
| | - Yessenia V S Ramos-Rivas
- Departamento de Ciencias Clinicas Facultad de Medicina, Universidad Nacional de la Amazonia Peruana, Iquitos, Peru
| |
Collapse
|
32
|
Ye Z, Yuan J, Hong D, Xu P, Liu W. Multimodal diagnostic models and subtype analysis for neoadjuvant therapy in breast cancer. Front Immunol 2025; 16:1559200. [PMID: 40170854 PMCID: PMC11958217 DOI: 10.3389/fimmu.2025.1559200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Background Breast cancer, a heterogeneous malignancy, comprises multiple subtypes and poses a substantial threat to women's health globally. Neoadjuvant therapy (NAT), administered prior to surgery, is integral to breast cancer treatment strategies. It aims to downsize tumors, optimize surgical outcomes, and evaluate tumor responsiveness to treatment. However, accurately predicting NAT efficacy remains challenging due to the disease's complexity and the diverse responses across different molecular subtypes. Methods In this study, we harnessed multimodal data, including proteomic, genomic, MRI imaging, and clinical information, sourced from multiple cohorts such as I-SPY2, TCGA-BRCA, GSE161529, and METABRIC. Post data preprocessing, Lasso regression was utilized for feature extraction and selection. Five machine learning algorithms were employed to construct diagnostic models, with pathological complete response (pCR) as the predictive endpoint. Results Our results revealed that the multi-omics Ridge regression model achieved the optimal performance in predicting pCR, with an AUC of 0.917. Through unsupervised clustering using the R package MOVICS and nine clustering algorithms, we identified four distinct multimodal breast cancer subtypes associated with NAT. These subtypes exhibited significant differences in proteomic profiles, hallmark cancer gene sets, pathway activities, tumor immune microenvironments, transcription factor activities, and clinical characteristics. For instance, CS1 subtype, predominantly ER-positive, had a low pCR rate and poor response to chemotherapy drugs, while CS4 subtype, characterized by high immune infiltration, showed a better response to immunotherapy. At the single-cell level, we detected significant heterogeneity in the tumor microenvironment among the four subtypes. Malignant cells in different subtypes displayed distinct copy number variations, differentiation levels, and evolutionary trajectories. Cell-cell communication analysis further highlighted differential interaction patterns among the subtypes, with implications for tumor progression and treatment response. Conclusion Our multimodal diagnostic model and subtype analysis provide novel insights into predicting NAT efficacy in breast cancer. These findings hold promise for guiding personalized treatment strategies. Future research should focus on experimental validation, in-depth exploration of the underlying mechanisms, and extension of these methods to other cancers and treatment modalities.
Collapse
Affiliation(s)
- Zheng Ye
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Jiaqi Yuan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
33
|
Alfawaz Altamimi AS, Arockia Babu M, Afzal M, Bishoyi AK, Roopashree R, Saini S, Sharma RSK, Pathak PK, Chauhan AS, Goyal K, Ali H, Khan NH, Balaraman AK. Exosomes derived from natural killer cells: transforming immunotherapy for aggressive breast cancer. Med Oncol 2025; 42:114. [PMID: 40100465 DOI: 10.1007/s12032-025-02647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Natural killer cell-derived exosomes (NK-Exos) hold great promise as immune modulators and immunotherapeutics against cancer due to their intrinsically latent anti-tumor effects. They use these nanosized vesicles to deliver cytotoxic molecules, such as perforin, granzymes, and miRNAs, directly to cancer cells to kill them, avoiding immune suppression. NK-Exos has particular efficacy for treating aggressive breast cancer by modulating the TME to activate the immune response and suppress immunosuppressive factors. Bioengineering advances have extended the therapeutic potential of NK-Exos, which permits precise tumor cell targeting and efficient delivery of therapeutic payloads, including small RNAs and chemotherapeutic agents. In engineered NK-Exos, sensitization of cancer cells to apoptosis, reduction of tumor growth, and resistance to drugs have been demonstrated to be highly effective. When combined, NK-Exos synergizes with radiotherapy, chemotherapy, or checkpoint inhibitors, enhancing therapeutic efficacy, and minimizing systemic toxicity. This review emphasizes the critical role of NK-Exos in breast cancer treatment, their integration into combination therapies, and the need for further research to overcome existing limitations and fully realize their clinical potential.
Collapse
Affiliation(s)
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - R S K Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Haider Ali
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor, Malaysia.
| |
Collapse
|
34
|
Čikeš Botić M, Marijan S, Radan M, Novak I, Glumac M, Pilkington LI, Odak Z, Barker D, Reynisson J, Čikeš Čulić V. Thieno[2,3- b]pyridines as a Novel Strategy Against Cervical Cancer: Mechanistic Insights and Therapeutic Potential. Int J Mol Sci 2025; 26:2651. [PMID: 40141292 PMCID: PMC11942470 DOI: 10.3390/ijms26062651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Cervical cancer is the fourth leading cause of cancer mortality in women worldwide, with limited therapeutic options for advanced or recurrent cases. In this study, the effects of a recent thieno[2,3-b]pyridine derivative, (E)-3-amino-5-(3-bromophenyl)acryloyl)-N-(3-chloro-2-methylphenyl)-6-methylthieno[2,3-b]pyridine-2-carboxamide (compound 1), on two cervical cancer cell lines, HeLa and SiHa, are investigated. Cytotoxicity was assessed by MTT assay, apoptosis rates were measured by flow cytometry, and metabolic profiling was performed by GC-MS. The study also examined the expression of eight glycosphingolipids (GSLs) in cancer stem cells (CSCs) and non-CSCs to assess glycophenotypic changes. Compound 1 showed significant cytotoxicity in both cell lines, with apoptosis identified as the primary mechanism of cell death. A significant reduction in the CSC population was observed, particularly in the SiHa cell line. Compound 1 treatment altered GSL expression and decreased GM2 levels in both CSCs and non-CSCs in the SiHa cell line and Gg3Cer levels in the HeLa cell line. Metabolic profiling identified 23 and 21 metabolites in the HeLa and SiHa cell lines, respectively, with significant differences in metabolite expression after treatment. These results underscore the potential of compound 1 as a promising therapeutic candidate for cervical cancer and warrant further investigation in preclinical and clinical settings.
Collapse
Affiliation(s)
- Monika Čikeš Botić
- Department of Gynecology and Obstetrics, University Hospital of Split, 21000 Split, Croatia;
| | - Sandra Marijan
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (S.M.); (V.Č.Č.)
| | - Mila Radan
- Department of Biochemistry, Faculty of Chemistry and Technology, University of Split, 21000 Split, Croatia;
| | - Ivana Novak
- Department of Immunology and Medical Genetics, School of Medicine, University of Split, 21000 Split, Croatia; (I.N.); (M.G.)
| | - Mateo Glumac
- Department of Immunology and Medical Genetics, School of Medicine, University of Split, 21000 Split, Croatia; (I.N.); (M.G.)
| | - Lisa I. Pilkington
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (L.I.P.); (D.B.)
- Te Pūnaha Matatini, Auckland 1042, New Zealand
| | - Zdravko Odak
- Department of Gynecology and Obstetrics, University Hospital of Split, 21000 Split, Croatia;
| | - David Barker
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand; (L.I.P.); (D.B.)
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6012, New Zealand
| | - Jóhannes Reynisson
- School of Allied Health Professions and Pharmacy, Keele University, Staffordshire ST5 5BG, UK;
| | - Vedrana Čikeš Čulić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (S.M.); (V.Č.Č.)
| |
Collapse
|
35
|
Gies S, Melchior P, Molnar I, Olmes G, Stroeder R, Tänzer T, Pohlers M, Schäfer M, Theobald L, Sester M, Solomayer EF, Walch-Rückheim B. PD-L1 + CD49f + CD133 + Circulating tumor cells predict outcome of patients with vulvar or cervical cancer after radio- and chemoradiotherapy. J Transl Med 2025; 23:321. [PMID: 40083005 PMCID: PMC11908062 DOI: 10.1186/s12967-025-06277-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/21/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Monitoring individual therapy responses of patients with cancer represents a major clinical challenge providing the basis to early identify metastases and cancer relapse. We previously demonstrated that radio- or chemoradiotherapy affects the systemic cellular milieu of patients with vulvar or cervical cancer and creates individual post-therapeutic environments associated with cancer relapse. Circulating tumor cells (CTCs) in the systemic milieu are related to metastases and relapse; however, their quantitative and phenotypic characteristics during therapy of patients with vulvar and cervical cancer are still unknown. METHODS In this prospective, longitudinal study, we verified the presence of CTCs via immunofluorescence and systemically characterized CTCs by flow cytometry from the blood of 40 patients with vulvar and 115 patients with cervical cancer receiving surgery, adjuvant radiotherapy (aRT), chemoradiotherapy (aCRT) or primary chemoradiotherapy (pCRT) and linked the presence of different CTC subpopulations with individual outcome of disease. RESULTS Pre-therapeutic cytokeratin+ CD45- CTC numbers significantly correlated with tumor FIGO stages, lymph node metastases and relapse. While surgery only did not significantly alter CTC occurrence, aRT and aCRT as well as pCRT differentially decreased or increased CTCs in patients with both tumor entities compared to baseline levels. Therapy-mediated increased CTC numbers were directly linked with subsequent cancer recurrence on follow-up. Phenotypic characterization of CTCs revealed enhanced expression of the stem cell marker CD133 as well as the integrin α6 (CD49f) after aRT, aCRT and pCRT. Furthermore, the aRT, aCRT and pCRT cohorts exhibited increased proportions of Programmed Cell Death Protein Ligand (PD-L1) expressing cells among post-therapeutic CTCs. Notably, post-therapeutic PD-L1+ CD49f+ CD133+ numbers ≥ 5/ml in patients with vulvar cancer and ≥ 2/ml in patients with cervical cancer were associated with reduced recurrence-free survival on follow-up. CONCLUSION Our study unravels individual therapy-induced changes in CTC phenotypic characteristics and occurrence in the patients' blood and their association with cancer relapse. Our results may help to explain differences in the individual courses of disease of patients with vulvar and cervical cancer and suggest PD-L1, CD49f and CD133 as targets for immunotherapy in vulvar and cervical cancer.
Collapse
Affiliation(s)
- Selina Gies
- Center of Human and Molecular Biology (ZHMB) Kirrbergerstraße, Institute of Virology, Saarland University, Building 47, D-66421, Homburg/Saar, Germany
- Experimental Gynaecological Oncology, Gynecology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Patrick Melchior
- Department of Radiation Oncology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Istvan Molnar
- Department of Obstetrics and Gynecology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Gregor Olmes
- Department of Obstetrics and Gynecology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Russalina Stroeder
- Department of Obstetrics and Gynecology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Tanja Tänzer
- Center of Human and Molecular Biology (ZHMB) Kirrbergerstraße, Institute of Virology, Saarland University, Building 47, D-66421, Homburg/Saar, Germany
| | - Maike Pohlers
- Center of Human and Molecular Biology (ZHMB) Kirrbergerstraße, Institute of Virology, Saarland University, Building 47, D-66421, Homburg/Saar, Germany
| | - Moritz Schäfer
- Center of Human and Molecular Biology (ZHMB) Kirrbergerstraße, Institute of Virology, Saarland University, Building 47, D-66421, Homburg/Saar, Germany
- Experimental Gynaecological Oncology, Gynecology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Laura Theobald
- Center of Human and Molecular Biology (ZHMB) Kirrbergerstraße, Institute of Virology, Saarland University, Building 47, D-66421, Homburg/Saar, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, Homburg/Saar, Germany
| | - Erich Franz Solomayer
- Department of Obstetrics and Gynecology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Barbara Walch-Rückheim
- Center of Human and Molecular Biology (ZHMB) Kirrbergerstraße, Institute of Virology, Saarland University, Building 47, D-66421, Homburg/Saar, Germany.
- Experimental Gynaecological Oncology, Gynecology, Faculty of Medicine, University of Augsburg, Augsburg, Germany.
| |
Collapse
|
36
|
Chen H, Ling R, Lai J, Liu Z, Wang Z, Yang H, Kong Y. CD44v6-mediated regulation of gastric cancer stem cells: a potential therapeutic target. Clin Exp Med 2025; 25:80. [PMID: 40069421 PMCID: PMC11897096 DOI: 10.1007/s10238-025-01611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
Gastric cancer is the fourth most common cause of cancer-related deaths globally. Cancer stem cells (CSCs) play an essential role in tumor initiation, development, and chemoresistance. However, the molecular mechanisms that regulate CSC traits in gastric cancer, particularly the role of CD44v6 as a key CSC marker, remain poorly understood. Here, we demonstrate that CD44v6 is markedly upregulated in gastric cancer tissues and correlates with poor prognosis. Functional assays, including colony formation, wound healing, proliferation, and apoptosis assays, show that CD44v6 enhances CSC characteristics, such as self-renewal, proliferation, migration, and cisplatin chemoresistance. CD44v6 knockdown effectively suppresses these aggressive phenotypes. Mechanistically, CD44v6 regulates the expression of key CSC markers, including CD24, CD133, EpCAM, as well as stemness-related transcription factors Oct-4 and Nanog. Additionally, CD44v6 enhances cell proliferation and drug resistance in both in vitro and in vivo experiments. Collectively, our findings highlight the significant role of CD44v6 in regulating gastric CSC traits, suggesting it's a potential as a biomarker and therapeutic target for improving gastric cancer treatment outcomes, particularly in overcoming chemoresistance.
Collapse
Affiliation(s)
- Hao Chen
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Ruoyu Ling
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Jiayu Lai
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Zhiqi Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, Guangdong Province, China.
| | - Hua Yang
- Department of Basic Medicine, School of Medicine, Foshan University, Foshan, 528225, Guangdong Province, China.
| | - Yi Kong
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology (SUAT), Shenzhen, 518107, Guangdong Province, China.
| |
Collapse
|
37
|
Chitren R, Gowda K, Amin S, Robertson GP, Jonnalagadda S, Budak-Alpdogan T, Pandey MK. Novel Pan-ALDH Inhibitor KS100 Effectively Targets ALDH+/CD138⁻ Stem-like Cells to Overcome Relapse in Multiple Myeloma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641909. [PMID: 40161728 PMCID: PMC11952311 DOI: 10.1101/2025.03.06.641909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Multiple myeloma (MM), a clonal plasma cell disorder is the second most frequent hematological malignancy in the United States. This malignancy is characterized by a series of symptoms such as bone lesions, hypercalcemia, renal failure, and anemia. The current clinical drugs in the market are successful in treating multiple myeloma patients into remission but does not address relapse where a more aggressive phenotype of the cancer remains untreatable. We hypothesize that a small subset of multiple myeloma stem-like cells (MMSLC's) that overexpress aldehyde dehydrogenases (ALDH + ) is the cause of the relapse. Overexpression of ALDH bolsters drug resistance via detoxification and stemness via the retinoic acid signaling pathway. The phenotype of MMSLC's is not yet known for certainty but there are a few well established markers such CD138 negative (CD138 neg ) cells that are known to overexpress ALDH. In this study, we target regular MM cells and bortezomib resistant ALDH + /CD138 neg MMSLC's with a novel, potent, pan-ALDH inhibitor, KS100. Here we report KS100 effectively lowered ALDH expression in regular and bortezomib resistant ALDH + /CD138 neg cells, MM cell viability as well as proteins associated with MMSLC's. Most importantly we showed that KS100 lowered ALDH + populations in regular, bortezomib resistant and CD138 neg cells via ALDEFLUOR™ assay.
Collapse
|
38
|
Thoenen E, Ranjan A, Parrales A, Nishikawa S, Dixon DA, Oka S, Iwakuma T. Suppression of stress granule formation is a vulnerability imposed by mutant p53. Nat Commun 2025; 16:2365. [PMID: 40064891 PMCID: PMC11894096 DOI: 10.1038/s41467-025-57539-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Missense mutations in the TP53 (p53) gene have been linked to malignant progression. However, our in-silico analyses reveal that hepatocellular carcinoma (HCC) patients with mutant p53 (mutp53) have better overall survival compared to those with p53-null (p53null) HCC, unlike other cancer types. Given the historical use of sorafenib (SOR) monotherapy for advanced HCC, we hypothesize that mutp53 increases sensitivity to SOR, a multikinase inhibitor that induces endoplasmic reticulum (ER) stress. Here we show that mutp53 inhibits stress granule (SG) formation by binding to an ER stress sensor, PKR-like ER kinase (PERK), and a key SG component, GAP SH3 domain-binding protein 1 (G3BP1), contributing to increased sensitivity of SG-competent cells and xenografts to ER stress inducers including SOR. Our study identifies a unique vulnerability imposed by mutp53, suggesting mutp53 as a biomarker for ER stress-inducing agents and highlighting the importance of SG inhibition for cancer treatment.
Collapse
Affiliation(s)
- Elizabeth Thoenen
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Atul Ranjan
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Alejandro Parrales
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Shigeto Nishikawa
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Dan A Dixon
- Department of Biochemistry and Molecular Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sugako Oka
- Faculty of Medical Science, Kyushu University, Fukuoka, Japan
| | - Tomoo Iwakuma
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA.
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
39
|
Liu C, Li Z, Cheng F, Li W, Li T. Research progress on molecular mechanism of liver metastasis of gastric cancer and treatment with traditional Chinese medicine. J Cancer 2025; 16:1944-1957. [PMID: 40092702 PMCID: PMC11905405 DOI: 10.7150/jca.105223] [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: 10/15/2024] [Accepted: 12/17/2024] [Indexed: 03/19/2025] Open
Abstract
Gastric cancer liver metastasis (GCLM) refers to the process of cancer cells from the stomach spreading to the liver, which is an important sign of the deterioration of gastric cancer (GC) and has a profound influence on the treatment and prognosis of patients. Once GC has liver metastasis, the treatment becomes more complex and challenging, which seriously affects the survival rate of patients with GC. Therefore, studying the mechanism and treatment of GCLM is extremely necessary. At present, the continuous research on GCLM has revealed that the mechanism of its occurrence and development involves the comprehensive effect of multiple targets and links. Traditional Chinese medicine (TCM) has the advantages of wide sources, excellent efficacy, and small toxicity and side effects, which have become the focus of current antitumor research. TCM, Chinese medicine monomers, or TCM compounds can inhibit the growth and metastasis of GC. In recent years, Chinese medicine has made substantial achievements in experimental research on the intervention of GCLM. This article reviews the progress of its intervention mechanism.
Collapse
Affiliation(s)
- Caiyue Liu
- Ningxia Medical University School of Traditional Chinese Medicine, Yinchuan, P. R. China
| | - Zheng Li
- Ningxia Medical University School of Traditional Chinese Medicine, Yinchuan, P. R. China
| | - Fane Cheng
- The First Clinical Medical College of Beijing University of Traditional Chinese Medicine, Beijing, P. R. China
| | - Weiqiang Li
- Ningxia Medical University School of Traditional Chinese Medicine, Yinchuan, P. R. China
- Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education (Ningxia Medical University), Yinchuan, P. R. China
- Ningxia medical university affiliated traditional Chinese medicine hospital, Yinchuan, P. R. China
| | - Tingting Li
- Ningxia Medical University School of Traditional Chinese Medicine, Yinchuan, P. R. China
- Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education (Ningxia Medical University), Yinchuan, P. R. China
| |
Collapse
|
40
|
Hwang J, Likasitwatanakul P, Deshmukh SK, Wu S, Kwon JJ, Toye E, Moline D, Evans MG, Elliott A, Passow R, Luo C, John E, Gandhi N, McKay RR, Heath EI, Nabhan C, Reizine N, Orme JJ, Domingo Domenech JM, Sartor O, Baca SC, Dehm SM, Antonarakis ES. Structurally Oriented Classification of FOXA1 Alterations Identifies Prostate Cancers with Opposing Clinical Outcomes and Distinct Molecular and Immunologic Subtypes. Clin Cancer Res 2025; 31:936-948. [PMID: 39745364 PMCID: PMC11873805 DOI: 10.1158/1078-0432.ccr-24-3471] [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: 10/18/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
PURPOSE Around 10% to 15% of prostate cancers harbor recurrent aberrations in the Forkhead Box A1 gene, FOXA1, whereby the alteration type and the effect on the forkhead (FKH) domain affect protein function. We developed a FOXA1 classification system to inform clinical management. EXPERIMENTAL DESIGN A total of 5,014 prostate cancer samples were examined using whole-exome and -transcriptome sequencing from the Caris Life Sciences database. We denoted class 1 FOXA1 alterations as missense and in-frame insertions/deletions with subclasses oriented with respect to the FKH domain. These were in the first part of the FKH domain [class 1A: amino acids (AA) 168-246], within the Wing2 region of FKH (class 1B: AA 247-269), or outside FKH (class 1C: AA 1-167, 270+). Two hotspot missense mutations at R219 were denoted class 2. Class 3 included predicted truncating mutations with subclasses partitioned based on the FKH domain (class 3A: AA 1-269 and class 3B: AA 270+). Class 4 represented FOXA1 amplifications. Real-world overall survival and therapy outcomes were determined from insurance claims. RESULTS FOXA1 alterations did not influence survival when considered in aggregate but had distinct prognostic effects when stratified by class. In primary prostate samples, class 1A alterations were associated with overall improved survival (HR, 0.57; P = 0.03); a similar trend was seen in metastatic biopsies with class 1B (HR, 0.84; P = 0.09). Conversely, in primary specimens, class 1C exhibited worse survival upon second-generation androgen receptor signaling inhibitor treatment (HR, 1.93; P < 0.001). Class 2 mutations (R219C/S) were enriched in neuroendocrine prostate cancers and were associated with overall poor survival (HR, 2.05; P < 0.001) and worse outcomes to first-line androgen-deprivation therapies (HR, 2.5; P < 0.001). Class 3A alterations indicated improved survival (HR, 0.70; P = 0.01), whereas class 3B alterations portended poor outcomes (HR, 1.50; P < 0.001). Amplifications (class 4) indicated poor outcomes in metastatic samples (HR, 1.48; P = 0.02). Molecularly, different FOXA1 alteration classes harbored distinct mutational and immunologic features as well as unique transcriptional programs. Finally, relative to European Americans, African Americans had increased class 1C alterations, whereas Asian/Pacific Islander patients had increased class 1B alterations. CONCLUSIONS FOXA1 alterations should not be interpreted in aggregate, as different classes are associated with divergent molecular features and clinical outcomes. Our revised classification schema facilitates clinical decision-making for patients with prostate cancer and uncovers important racial differences.
Collapse
Affiliation(s)
- Justin Hwang
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Pornlada Likasitwatanakul
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Dana Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Sharon Wu
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Jason J. Kwon
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Eamon Toye
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Moline
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Mark G. Evans
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Andrew Elliott
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Rachel Passow
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Christine Luo
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Emily John
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Nishant Gandhi
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Rana R. McKay
- University of California San Diego, San Diego, California
| | | | - Chadi Nabhan
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | | | | | | | | | | | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Emmanuel S. Antonarakis
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
41
|
Alshammari QA, Alshammari SO, Alshammari A, Alfarhan M, Baali FH. Unraveling the mechanisms of glioblastoma's resistance: investigating the influence of tumor suppressor p53 and non-coding RNAs. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2569-2585. [PMID: 39476245 DOI: 10.1007/s00210-024-03564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/22/2024] [Indexed: 01/04/2025]
Abstract
Glioblastoma (GB) is one of the most fatal CNS malignancies, and its high resistance to therapy and poor outcomes have made it one of the primary challenges in oncology. Resistance to standard therapy, i.e., radio-chemotherapy with temozolomide, is one of the principal causes of the poor prognostic outcomes of GB. Finding the molecular basis of GB resistance to therapy is key to creating effective solution approaches. The general problem of GB resistance is supervised by cancer suppressive protein, p53, and has become a very special interest in molecular research in recent decades. The principal aim of this manuscript is to perform a comprehensive survey on the complex network of interactions developed by p53 with non-coding RNAs (ncRNA) in the context of GB resistance. The present article details the functional aspects of p53 as a cellular stress response protein, including its roles in apoptosis, cell cycle regulation, and DNA repair in glioblastoma (GB), along with the disruption of p53 and its involvement in chemoresistance (CR). It also highlights several classes of ncRNAs, namely microRNAs, long ncRNAs, and circular RNAs, that manipulate p53 signaling in GB-CR. The article likewise explains how disruption in the expression of these ncRNAs can promote GB-CR and how it interacts with essential cellular functions, such as proliferation, apoptosis, and DNA repair. The manuscript also describes the potential of targeting p53 and ncRNAs with their diagnostic and prognostic potential as novel promising therapeutics for GB. Nevertheless, ncRNA-based biomarkers still present challenges for their suitability in GB resistance. However, modern research continues to discover novel prediction targets, potentially enhancing patient outcomes and therapeutic options. Therefore, the neutralization of this intricate regulatory network of GB resistance might have a primary clinical effect in fighting GB resistance therapy and thus might lead to a substantial increase in patient survival and quality of life.
Collapse
Affiliation(s)
- Qamar A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, College of Pharmacy, Northern Border University, 76321, Rafha, Saudi Arabia
| | - Abdulkarim Alshammari
- Department of Pharmacy Practice, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Moaddey Alfarhan
- Department of Clinical Practice, College of Pharmacy, Jazan University, 45142, Jazan, Jizan, Saudi Arabia
| | - Fahad Hassan Baali
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| |
Collapse
|
42
|
Santos LS, Tonel MZ, Martins MO, dos Santos CL. Theoretical Exploration of Chitosan Nanoparticles Associated with Platinum Compounds for Cancer Treatment: Insights from DFT and Molecular Docking Analyses. BIONANOSCIENCE 2025; 15:79. [DOI: 10.1007/s12668-024-01728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 01/04/2025]
|
43
|
Atalaya-Marin N, Goñas M, Tineo D, Chuquibala-Checan B, Arce-Inga M, Tarrillo E, Alvarez-Robledo YA, Tafur-Culqui J, Cabrera-Hoyos H, Gómez-Fernández D. Integrating remote sensing and in-situ data to determine climate diversity and variability in cocoa systems in the provinces of Jaén and San Ignacio, Cajamarca (NW Perú). TREES, FORESTS AND PEOPLE 2025; 19:100749. [DOI: 10.1016/j.tfp.2024.100749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
44
|
Uygun DFK, Uygun V, Başaran A, Kocatepe G, Kazlı T, Bingöl A. High malignancy rate in IgE-deficient children. Int J Cancer 2025; 156:964-968. [PMID: 39369446 PMCID: PMC11701383 DOI: 10.1002/ijc.35213] [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: 06/22/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
Recent epidemiological studies have increasingly highlighted the antitumor efficacy of IgE owing to the increased malignancy rate in IgE-deficient patients. The purpose of this study, the largest for children, was to determine whether malignant diagnoses in children are associated with IgE deficiency (IgE <2.5 kIU/L). A total of 6821 pediatric patients were reviewed, focusing on patients with IgE below 2.5 kIU/L (n = 599). The causes of IgE testing were evaluated by categorizing them as having cancer, allergies, suspected or diagnosed immunodeficiency, and other conditions. In all but one patient with malignancy, IgE levels were measured after the diagnosis of the disease. Malignancies were observed much more frequently in the low IgE group than in the normal group (10/599, 1.7% and 7/6222, 0.11%; OR = 15.07; 95% CI: 5.72-39.75; p <.0001). According to our analysis, 70% of the patients had leukemia/lymphoma, which is consistent with studies showing that hematologic malignancies are the most frequent cancers linked to IgE deficiency. No increase in the prevalence of cancer was observed in IgE-deficient patients with suspected or diagnosed immunodeficiency. In conclusion, we observed a higher rate of previous malignancy (particularly hematologic cancer) in children with low serum IgE levels. Larger investigations would offer insightful information about the function of low IgE levels in predicting malignancy risk and improving the present diagnostic procedures.
Collapse
Affiliation(s)
| | - Vedat Uygun
- Department of Pediatric Bone Marrow Transplantation Unitİstinye University School of Medicine, MedicalPark Antalya HospitalAntalyaTurkey
| | - Ayşen Başaran
- Pediatric Pulmonology DepartmentAkdeniz University School of MedicineAntalyaTurkey
| | - Gaye Kocatepe
- Pediatric Allergy‐Immunology DepartmentAkdeniz University School of MedicineAntalyaTurkey
| | - Tuğba Kazlı
- Pediatric Pulmonology DepartmentAkdeniz University School of MedicineAntalyaTurkey
| | - Ayşen Bingöl
- Pediatric Allergy‐Immunology DepartmentAkdeniz University School of MedicineAntalyaTurkey
| |
Collapse
|
45
|
Kundu S, Jit BP, Sharma A. Pan-cancer TCGA analysis reveals the potential involvement of B-type lamins in dysregulating chromosome segregation in human cancer. Mamm Genome 2025; 36:230-249. [PMID: 39592474 DOI: 10.1007/s00335-024-10086-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024]
Abstract
Lamins play a crucial role in maintaining nuclear structure and function. Our study investigates the expression patterns and clinical implications of B-type lamins with a special focus on lamin B2 across various cancer types using comprehensive RNA sequencing datasets. We found that high expression levels of lamin B1 and lamin B2 are associated with decreased overall and disease-free survival in cancer. This association is further pronounced when both lamins are co-expressed, indicating a compounded negative impact on patient prognosis. Additionally, we highlighted the relationship between B-type lamins and the tumor microenvironment. Lamin B1 mRNA expression shows a strong positive correlation with activated CD4+ T-cells and type 2 T-helper cells (Th2), suggesting its role in immune cell infiltration and response within tumors. Lamin B2 expression also correlates moderately with these immune cells, indicating a potential but lesser role in modulating the immune landscape. Notably, the epigenetic state of lamin B1 significantly affects the tumor microenvironment, suggesting a dual role in structural integrity and immune modulation. We have identified 9 lamin B2 interacting proteins that are co-expressed with B-type lamins in cancerous conditions and modulate cytokinesis and cell division pathways during tumorigenesis. Furthermore, we have identified specific molecular targets of B-type lamins that co-express with them in a range of human cancers and are potentially involved in dysregulating chromosome segregation and mRNA binding. Overexpression of these targets alongside B-type lamins correlates with poor prognosis in multiple cancers. These findings underscore the potential of B-type lamins as biomarkers for poor prognosis and as targets for cancer therapies.
Collapse
Affiliation(s)
- Subhadip Kundu
- Laboratory of Chromatin and Cancer Epigenetics, Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Bimal Prasad Jit
- Laboratory of Chromatin and Cancer Epigenetics, Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ashok Sharma
- Laboratory of Chromatin and Cancer Epigenetics, Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
- National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, Haryana, India.
| |
Collapse
|
46
|
Sadahiro Y, Okubo M, Hitora Y, Hitora-Imamura N, Kotani S, Tsukamoto S. Pestones A and B from a Fungus Pestalotiopsis sp. Bound to Mutant p53 and Changed Its Conformation. JOURNAL OF NATURAL PRODUCTS 2025; 88:546-553. [PMID: 39952908 DOI: 10.1021/acs.jnatprod.4c01440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
Oncogenic mutant p53 is one of the targets for cancer therapy, and the development of anticancer drugs that reactivate mutant p53 is a promising strategy. The extract of fungus Pestalotiopsis sp. changed mutant p53 to wild-type-like p53 in Saos-2 (p53R175H) cells, as shown by fluorescent immunostaining, and bioassay-guided purification of the extract afforded new dimeric epoxyquinoids, pestones A and B (1 and 2), and a known compound, rosnecatrone (3). The relative and absolute configurations of 1 and 2 were determined based on the spectroscopic data and semisynthesis from 3. Compounds 1 and 2 altered the conformation of mutant p53 in Saos-2 (p53R175H) cells, as shown by immunofluorescence staining. The cellular thermal shift assay analysis showed that 1 increased the thermostability of mutant p53 in Saos-2 (p53R175H) cells, suggesting the direct binding of 1 to mutant p53. Compounds 1 and 2 exhibited cytotoxic activities against Saos-2 (p53R175H) cells with IC50 values of 1.0 and 1.1 μM, respectively. Compound 1 was found to induce apoptosis in Saos-2 (p53R175H) cells by flow cytometry analysis and decreased tumor growth in vivo using a mouse model with HuCCT1 (p53R175H) cells.
Collapse
Affiliation(s)
- Yusaku Sadahiro
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Misaki Okubo
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yuki Hitora
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Natsuko Hitora-Imamura
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Shunsuke Kotani
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Sachiko Tsukamoto
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| |
Collapse
|
47
|
Masuda Y, Sadato D, Toya T, Hosoda Y, Hirama C, Shimizu H, Najima Y, Harada H, Harada Y, Doki N. Transplantation outcomes of TP53-mutant AML and MDS: a single transplantation center experience of 63 patients. Int J Hematol 2025:10.1007/s12185-025-03951-z. [PMID: 40011351 DOI: 10.1007/s12185-025-03951-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
Allogeneic hematopoietic stem cell transplantation is recommended for TP53-mutant acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) despite a high relapse rate and poor survival. To understand TP53 alterations on a molecular level and define stratified prognostic outcomes following transplantation, we performed targeted next-generation sequencing on 63 patients who underwent transplantation for TP53-mutant AML/MDS and profiled their molecular spectrum. Sixty-eight TP53 mutations were detected, with a median variant allele frequency of 46.8%. Copy number alterations at the TP53 locus were present in 19 patients (30%). Complex karyotype was detected in 48 patients (76%) and was significantly associated with larger TP53 clone size, bi-allelic status, and the absence of concurrent mutations, reflecting the high TP53 mutational burden. Specifically, 51 patients (81%) with the dominant TP53 clone greatly overlapped with those with the complex karyotype. Multivariable overall survival (OS) analysis identified AML (hazard ratio [HR], 2.51; P = 0.03) and TP53 clonal dominance (HR, 5.30; P = 0.002) as prognostic factors. One-year OS was worse in AML with the dominant TP53 clone than in others (13% vs 61%; P < 0.001). Our results underscore the utility of mutational profile-guided risk stratification in patients with TP53-mutant AML/MDS, and could aid in transplantation-related decision-making.
Collapse
Affiliation(s)
- Yasutaka Masuda
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Daichi Sadato
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Takashi Toya
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan.
| | - Yuzuru Hosoda
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Chizuko Hirama
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hiroaki Shimizu
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Hironori Harada
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuka Harada
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| |
Collapse
|
48
|
Feng K, Li J, Li J, Li Z, Li Y. Prognostic implications of ERLncRNAs in ccRCC: a novel risk score model and its association with tumor mutation burden and immune microenvironment. Discov Oncol 2025; 16:225. [PMID: 39985635 PMCID: PMC11846825 DOI: 10.1007/s12672-025-01870-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/03/2025] [Indexed: 02/24/2025] Open
Abstract
INTRODUCTION/BACKGROUND The specific role of efferocytosis-related long noncoding RNAs (ERLncRNAs) in Clear Cell Renal Cell Carcinoma (ccRCC) has not been thoroughly examined. This study aims to identify and validate a signature of ERLncRNAs for prognostic prediction and characterization of the immune landscape in individuals with ccRCC. MATERIALS AND METHODS Analysis of ccRCC samples was conducted by utilizing clinical and RNA sequencing information obtained from The Cancer Genome Atlas (TCGA). Pearson correlation analysis was utilized to identify lncRNAs associated with efferocytosis, which was then used to create a new prognostic model through univariate Cox regression, Least Absolute Shrinkage and Selection Operator (LASSO) regression, and stepwise multivariate Cox analysis. In order to investigate the biological significance, we performed a functional enrichment analysis to assess how well the model predicts outcomes. Differences in the immune landscape were observed through a comparison of immune cell infiltration, tumor mutational burden (TMB), and tumor microenvironment (TME) characteristics. Following this, drug sensitivity analysis was conducted. RESULTS This led to the identification of a unique signature consisting of seven ERLncRNAs (LINC01615, RUNX3-AS1, FOXD2-AS1, AC002070.1, LINC02747, LINC00944, and AC092296.1). Model performance was measured by Kaplan-Meier curves and receiver operating characteristic (ROC) curves. The nomogram and C-index provided additional validation of the strong correlation between the risk signature and clinical decision-making. CONCLUSION On the whole, our innovative signature exhibits potential for prognostic prediction and assessment of immunotherapeutic response in patients with ccRCC.
Collapse
Affiliation(s)
- Kunlun Feng
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, Shandong, China
| | - Jingxiang Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianye Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhichao Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Yahui Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
49
|
Cao B, Liu M, Xiao Z, Leng D, Zhou Y, Zhang Z, Wang L, Huang X, Ni Q, Cheng W, Assaraf YG, Zhao Q, Shen J, Zhu K. CV1-secreting sCAR-T cells potentiate the abscopal effect of microwave ablation in heterogeneous tumors. Cell Rep Med 2025; 6:101965. [PMID: 39970874 PMCID: PMC11866491 DOI: 10.1016/j.xcrm.2025.101965] [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: 05/24/2024] [Revised: 10/01/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
Microwave ablation (MWA) triggers a weak systemic immune response that leads to the abscopal regression of distant metastases while killing local tumors, known as the abscopal effect. Combining MWA with chimeric antigen receptor (CAR)-T cells demonstrates promise in enhancing the abscopal effect in antigen-homogeneous tumors. However, the loss of the antigen recognized by CAR or intrinsic antigenic heterogeneity in solid tumors poses a major obstacle. SIRPα variant (CV1)-secreting CAR-T (sCAR-T) cells elicit an abscopal effect on distant tumors with antigen heterogeneity in mice receiving local MWA. Mechanistically, sCAR-T cells can locally eliminate antigen-positive tumors and secrete CV1, whereas the secreted CV1 can activate macrophages that migrate to non-ablated tumor sites in response to post-MWA chemokines, eliciting a macrophage-dependent abscopal effect that enables phagocytosis of antigen-heterogeneous cancer cells. This macrophage-dependent abscopal effect instigated by MWA and sCAR-T cells offers a clinically translatable strategy in metastatic solid tumors with antigen heterogeneity.
Collapse
Affiliation(s)
- Bihui Cao
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China; Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Manting Liu
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Zecong Xiao
- Nanomedicine Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Dongliang Leng
- MoE Frontiers Science Center for Precision Oncology, Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Yubo Zhou
- Department of Library, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhenfeng Zhang
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Lu Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xinkun Huang
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Wei Cheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China.
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| |
Collapse
|
50
|
Lim JU, Jung J, Kim YW, Kim CY, Lee SH, Park DW, Choi SI, Ji W, Yeo CD, Lee SH. Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges. Biomedicines 2025; 13:470. [PMID: 40002883 PMCID: PMC11852785 DOI: 10.3390/biomedicines13020470] [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: 01/10/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have transformed the treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer. However, treatment resistance remains a major challenge in clinical practice. The tumor microenvironment (TME) is a complex system composed of tumor cells, immune and non-immune cells, and non-cellular components. Evidence indicates that dynamic changes in TME during TKI treatment are associated with the development of resistance. Research has focused on identifying how each component of the TME interacts with tumors and TKIs to understand therapeutic targets that could address TKI resistance. In this review, we describe how TME components, such as immune cells, fibroblasts, blood vessels, immune checkpoint proteins, and cytokines, interact with EGFR-mutant tumors and how they can promote resistance to TKIs. Furthermore, we discuss potential strategies targeting TME as a novel therapeutic approach.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon Wook Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Chi Young Kim
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sang Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Institute of Chest Diseases, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Won Park
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea;
| | - Sue In Choi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Wonjun Ji
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 44610, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03083, Republic of Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|