1
|
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
|
2
|
Abusharieh E, Aslam N, Zihlif MA, Bustanji Y, Wehaibi S, Abuarqoub D, Shahin D, Saadeh H, Barham R, Awidi AS. In vitro investigation of epigenetic regulators related to chemo-resistance and stemness of CD133 +VE cells sorted from U87MG cell line. Gene 2025; 956:149432. [PMID: 40157620 DOI: 10.1016/j.gene.2025.149432] [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: 09/17/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Glioblastoma (GBM) is the most common and malignant adult primary brain tumor with frequent relapse and resistance to therapies. Glioma stem cells, a rare population, is thought to be the reason behind the treatment's failure. It is imperative to investigate the disease mechanisms and identify the biomarkers by which glioma stem cells would contribute to treatment relapse and resistance to already available chemotherapeutic agents. The CD133+VE cells were isolated from U87MG cell line and characterized by morphological features, cell viability, self-renewal efficiency, migration potential and karyotyping. Doxorubicin Cisplatin, Irinotecan, Etoposide and Temozolomide were used to determine the anti-proliferative effect on CD133+VE cells. Confocal microcopy was used to localize the chemotherapeutic agents in the CD133+VE cells. In quest of epigenetic biomarkers, RNA sequencing was performed to find the role of lncRNAs in stemness and resistance to therapies. U87cell line and CD133-VE cells were kept as controls for all the experiments. It was found that CD133+VEcells were highly proliferative with increased migration potential, elevated IC50 values against chemotherapeutic agents and showed distinct karyotyping related to pluripotency. Chemotherapeutic agent such as Doxorubicin was localized outside the nucleus revealing the drug resistance as evident by confocal microscopy. RNA sequencing revealed 126 differentially expressed lncRNAs (DELs) in CD133+VEcells among which lncRNA LOXL1-AS1 was highly upregulated and lncRNA PAX8-AS1 was significantly downregulated. These lncRNAs has been reported to be related to drug resistance, migration and epithelial- to- mesenchymal transmission (EMT), self-renewal and stemness properties contributing to poor prognosis and disease relapse.
Collapse
Affiliation(s)
- Elham Abusharieh
- Department of Pharmaceutical Science, Faculty of Pharmacy, Al-zaytoonah University of Jordan, Amman 11733, Jordan; Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; Department of Clinical Pharmacy and Biopharmaceutics, Faculty of Pharmacy, The University of Jordan, Amman, Jordan.
| | - Nazneen Aslam
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Malek A Zihlif
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Yasser Bustanji
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Clinical Pharmacy and Biopharmaceutics, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Suha Wehaibi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Duaa Abuarqoub
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra. Amman 11196, Jordan
| | - Diana Shahin
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Heba Saadeh
- Department of Computer Science, KASIT, The University of Jordan, Amman, 11942 Jordan
| | - Raghad Barham
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Abdalla S Awidi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; Department of Hematology and Oncology, Jordan University Hospital, The University of Jordan, Amman 11942, Jordan.
| |
Collapse
|
3
|
Pagliari F, Tirinato L, Di Fabrizio E. Raman spectroscopies for cancer research and clinical applications: a focus on cancer stem cells. Stem Cells 2025; 43:sxae084. [PMID: 39949042 DOI: 10.1093/stmcls/sxae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/20/2024] [Indexed: 04/23/2025]
Abstract
Over the last 2 decades, research has increasingly focused on cancer stem cells (CSCs), considered responsible for tumor formation, resistance to therapies, and relapse. The traditional "static" CSC model used to describe tumor heterogeneity has been challenged by the evidence of CSC dynamic nature and plasticity. A comprehensive understanding of the mechanisms underlying this plasticity, and the capacity to unambiguously identify cancer markers to precisely target CSCs are crucial aspects for advancing cancer research and introducing more effective treatment strategies. In this context, Raman spectroscopy (RS) and specific Raman schemes, including CARS, SRS, SERS, have emerged as innovative tools for molecular analyses both in vitro and in vivo. In fact, these techniques have demonstrated considerable potential in the field of cancer detection, as well as in intraoperative settings, thanks to their label-free nature and minimal invasiveness. However, the RS integration in pre-clinical and clinical applications, particularly in the CSC field, remains limited. This review provides a concise overview of the historical development of RS and its advantages. Then, after introducing the CSC features and the challenges in targeting them with traditional methods, we review and discuss the current literature about the application of RS for revealing and characterizing CSCs and their inherent plasticity, including a brief paragraph about the integration of artificial intelligence with RS. By providing the possibility to better characterize the cellular diversity in their microenvironment, RS could revolutionize current diagnostic and therapeutic approaches, enabling early identification of CSCs and facilitating the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Francesca Pagliari
- Division of Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Luca Tirinato
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Enzo Di Fabrizio
- PolitoBIOMed Lab DISAT Department, Polytechnic University of Turin, 10129 Turin, Italy
| |
Collapse
|
4
|
Li H, Jia F, Wang X, Yang T, Wang JH. Efficient and Discriminative Isolation of Circulating Cancer Stem Cells and Non-Stem-like Circulating Tumor Cells Using a Click-Handle-Loaded M13 Phage-Based Surface. Anal Chem 2025; 97:8080-8087. [PMID: 40192481 DOI: 10.1021/acs.analchem.5c00924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Circulating tumor cells (CTCs) are crucial for cancer research and clinical applications, with circulating cancer stem cells (cCSCs) being a rare but key subpopulation responsible for metastasis, recurrence, and therapy resistance. Current limitations in efficiently isolating these cells, particularly distinguishing cCSCs from non-stem-like CTCs (nsCTCs), hinder our understanding of cancer progression and precision medicine strategies. Herein, we developed a novel CTC isolation approach that integrates cell metabolic chemical tagging with a click-handle-loaded M13 phage-based surface (CHPhace). The multivalent nature of flexible M13 nanofibers, featuring thousands of modification sites for click reactions, significantly enhances CTC capture across diverse tumor types. Leveraging the unique slow-cycling characteristic of cCSCs, CHPhace demonstrated selective cCSCs isolation through metabolic labeling and demetabolism processes. The robust performance of CHPhace allows efficient isolation of both cCSCs and nsCTCs from complex blood sample matrices, achieving capture efficiencies exceeding 80%. This approach represents a promising tool for advancing our understanding of cancer progression and enhancing precision in clinical diagnosis and cancer prognosis.
Collapse
Affiliation(s)
- Huida Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Fengting Jia
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xin Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Ting Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
5
|
Wang J, Liao J, Cheng Y, Chen M, Huang A. LAPTM4B enhances the stemness of CD133 + liver cancer stem-like cells via WNT/β-catenin signaling. JHEP Rep 2025; 7:101306. [PMID: 40171299 PMCID: PMC11960653 DOI: 10.1016/j.jhepr.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 04/03/2025] Open
Abstract
Background & Aims Lysosome-associated protein transmembrane 4β (LAPTM4B) is an oncogene implicated in the malignant progression of hepatocellular carcinoma (HCC). Previous research established a strong association between LAPTM4B and HCC stemness. However, specific mechanisms by which LAPTM4B regulates and maintains the stemness of liver cancer stem cells remain unclear. Therefore, we investigated the effects of LAPTM4B on the stemness regulation of cluster of differentiation 133 (CD133)+ liver cancer stem-like cells (CSLCs). Methods We used RNA interference and overexpression techniques in both in vitro and in vivo models. The involvement of LAPTM4B in wingless/integrated (WNT)/β-catenin signaling was examined through western blotting, immunofluorescence, and immunoprecipitation. The impact of LAPTM4B on β-catenin phosphorylation and ubiquitination was analyzed to elucidate its role in promoting stemness. Clinical relevance was evaluated in an in-house cohort of 105 specimens from patients with HCC through immunohistochemical and microarray analysis, enabling investigation of correlations with clinical outcomes. Results LAPTM4B promoted the self-renewal ability, chemoresistance, and tumorigenicity of CD133+ CSLCs. Mechanistically, aberrant LAPTM4B upregulation facilitated β-catenin nuclear translocation (nucleocytoplasmic separation assay, p <0.001) and inhibited its phosphorylation (p <0.01). In addition, LAPTM4B interacts with the deubiquitinating enzymes ubiquitin carboxyl-terminal hydrolase (USP)-1 and USP14, reducing β-catenin ubiquitination. Furthermore, patients with high LAPTM4B and β-catenin expression had markedly shorter 3-year overall survival rate (42.9% vs. 74.4%; hazard ratio, 5.174; 95% CI 2.280-11.741, p <0.001). Conclusions LAPTM4B promotes CD133+ CSLC stemness by activating WNT/β-catenin signaling by inhibiting β-catenin phosphorylation and ubiquitination degradation. The role of LAPTM4B in regulating WNT/β-catenin signaling suggests that LAPTM4B serves as a therapeutic target for impairing HCC stemness and progression. Impact and implications LAPTM4B contributes significantly to CD133+ CSLC stemness and inhibits β-catenin phosphorylation and ubiquitination degradation, activating WNT/β-catenin signaling. WNT inhibitors suppress LAPTM4B-induced CD133+ CSLC stemness. Thus, targeting the LAPTM4B-WNT/β-catenin axis could improve antitumor efficacy.
Collapse
Affiliation(s)
- Jiahong Wang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
| | - Jianping Liao
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| | - Ye Cheng
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Diagnostical Pathology Center, Fujian Medical University, Fuzhou, Fujian 350004, China
| | - Meirong Chen
- Department of Pathology, Quanzhou Maternity and Children’s Hospital, Quanzhou, Fujian 362000, China
| | - Aimin Huang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
| |
Collapse
|
6
|
Varlamova EG. Roles of selenium-containing glutathione peroxidases and thioredoxin reductases in the regulation of processes associated with glioblastoma progression. Arch Biochem Biophys 2025; 766:110344. [PMID: 39956249 DOI: 10.1016/j.abb.2025.110344] [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: 10/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Glioblastoma remains the most common and aggressive primary tumor of the central nervous system in adults. Current treatment options include standard surgical resection combined with radiation/chemotherapy, but such protocol most likely only delays the inevitable. Therefore, the problem of finding therapeutic targets to prevent the occurrence and development of this severe oncological disease is currently acute. It is known that the functions of selenoproteins in the regulation of carcinogenesis processes are not unambiguous. Either they exhibit cytotoxic activity on cancer cells, or cytoprotective. A special place in the progression of oncological diseases of various etiologies is occupied by proteins of the thioredoxin and glutathione systems. These are two cellular antioxidant systems that regulate redox homeostasis, counteracting the increased production of reactive oxygen species in cells. The review reflects the latest data on the role of key enzymes of these redox systems in the regulation of processes associated with the progression of glioblastoma. A thorough consideration of these issues will expand fundamental knowledge about the functions of selenium-containing thioredoxin reductases and glutathione peroxidases in the therapy of glioblastomas and provide an understanding of the prospects for the treatment of this aggressive oncological disease.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", St. Institutskaya 3, Pushchino, 142290, Russia.
| |
Collapse
|
7
|
Li S, Gao S, Qin L, Ding C, Qu J, Cui Y, Qiang L, Yin S, Zheng X, Meng H. Micropapillary structure: A natural tumor collective invasion model with enhanced stem-like properties. Cancer Sci 2025; 116:308-321. [PMID: 39568148 PMCID: PMC11786311 DOI: 10.1111/cas.16396] [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: 06/24/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Cancer stem cells aggregate to form clusters, which have enhanced stem-like properties and metastasis potential. However, the molecular mechanisms underlying the formation of cancer stem cell cluster-like structures with acquisition of stronger invasion and metastasis abilities remain unclear. Micropapillary carcinoma (MPC) is a subpopulation of small, merulioid, inverted, nonfibrous vascular clusters floating in the stroma present in a range of solid malignant tumors and characterized by frequent vascular/lymphatic vessel invasion and lymph node metastasis. Our results showed that these cell clusters exhibit a stem cell phenotype, supporting the premise that MPC may serve as a promising solid tumor model for studying invasion and metastasis of cancer stem cell clusters. In this review, we discuss the latest advances in MPC research and targeted therapy, focusing on analysis of their stem-like characteristics, mapping their multiomics characteristics, and elucidating the vascular and immune microenvironment of MPC. The existing MPC organoid model was employed to explore potential breakthroughs in targeted therapy and immunotherapy for cancer stem cell clusters.
Collapse
Affiliation(s)
- Sisi Li
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Shuangshu Gao
- Department of PathologyHarbin Medical UniversityHarbinChina
| | - Ling Qin
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Caixia Ding
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Jinghui Qu
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Yifei Cui
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Lixia Qiang
- Department of Respiratory MedicineThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Shengjie Yin
- Department of Medical OncologyMunicipal Hospital of ChifengChifengChina
| | - Xiaoyu Zheng
- Department of AnesthesiologyHarbin Medical University Cancer HospitalHarbinChina
| | - Hongxue Meng
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| |
Collapse
|
8
|
Ragab EM, Gamal DME, El-Najjar FF, Elkomy HA, Ragab MA, Elantary MA, Basyouni OM, Moustafa SM, El-Naggar SA, Elsherbiny AS. New insights into Notch signaling as a crucial pathway of pancreatic cancer stem cell behavior by chrysin-polylactic acid-based nanocomposite. Discov Oncol 2025; 16:107. [PMID: 39891818 PMCID: PMC11787125 DOI: 10.1007/s12672-025-01846-3] [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: 10/31/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Pancreatic cancer is an extremely deadly illness for which there are few reliable treatments. Recent research indicates that malignant tumors are highly variable and consist of a tiny subset of unique cancer cells, known as cancer stem cells (CSCs), which are responsible for the beginning and spread of tumors. These cells are typically identified by the expression of specific cell surface markers. A population of pancreatic cancer stem cells with aberrantly active developmental signaling pathways has been identified in recent studies of human pancreatic tumors. Among these Notch signaling pathway has been identified as a key regulator of CSCs self-renewal, making it an attractive target for therapeutic intervention. Chrysin-loaded polylactic acid (PLA) as polymeric nanoparticles systems have been growing interest in using as platforms for improved drug delivery. This review aims to explore innovative strategies for targeted therapy and optimized drug delivery in pancreatic CSCs by manipulating the Notch pathway and leveraging PLA-based drug delivery systems. Furthermore, we will assess the capability of PLA nanoparticles to enhance the bioavailability and effectiveness of gemcitabine in pancreatic cancer cells. The insights gained from this review have the potential to contribute to the development of novel treatment approaches that combine targeted therapy with advanced drug delivery utilizing biodegradable polymeric nanoparticles.
Collapse
Affiliation(s)
- Eman M Ragab
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doaa M El Gamal
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Fares F El-Najjar
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Hager A Elkomy
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud A Ragab
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mariam A Elantary
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Omar M Basyouni
- Chemistry/Zoology Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sherif M Moustafa
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Shimaa A El-Naggar
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Abeer S Elsherbiny
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
9
|
Jiang X, Nik Nabil WN, Ze Y, Dai R, Xi Z, Xu H. Unlocking Natural Potential: Antibody-Drug Conjugates With Naturally Derived Payloads for Cancer Therapy. Phytother Res 2025; 39:789-874. [PMID: 39688127 DOI: 10.1002/ptr.8407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Natural compound-derived chemotherapies remain central to cancer treatment, however, they often cause off-target side effects that negatively impact patients' quality of life. In contrast, antibody-drug conjugates (ADCs) combine cytotoxic payloads with antibodies to specifically target cancer cells. Most approved and clinically investigated ADCs utilize naturally derived payloads, while those with conventional synthetic molecular payloads remain limited. This review focuses on approved ADCs that enhance the efficacy of naturally derived payloads by linking them with antibodies. We provide an overview of the core components of ADCs, their working mechanisms, and FDA-approved ADCs featuring naturally derived payloads, such as calicheamicin, camptothecin, dolastatin 10, maytansine, pyrrolbenzodiazepine (PBD), and the immunotoxin Pseudomonas exotoxin A. This review also explores recent clinical advancements aimed at broadening the therapeutic potential of ADCs, their applicability in treating heterogeneously composed tumors and their potential use beyond oncology. Additionally, this review highlights naturally derived payloads that are currently being clinically investigated but have not yet received approval. By summarizing the current landscape, this review provides insights into promising avenues for exploration and contributes to the refinement of treatment protocols for improved patient outcomes.
Collapse
Affiliation(s)
- Xue Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- National Pharmaceutical Regulatory Agency, Ministry of Health, Selangor, Malaysia
| | - Yufei Ze
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
10
|
Tian J, Li Y, Tong Y, Zhang Y, Zhao T, Kang Y, Bi Q. Uridine-cytidine kinase 2 is correlated with immune, DNA damage repair and promotion of cancer stemness in pan-cancer. Front Oncol 2025; 15:1503300. [PMID: 39931080 PMCID: PMC11807824 DOI: 10.3389/fonc.2025.1503300] [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: 09/28/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
Background UCK2 (Uridine-Cytidine Kinase 2) is a promising prognostic marker for malignant tumors, but its association with immune infiltration and cancer stemness in pan-cancer remains to be fully understood. we find that gene UCK2 is closed related to RNA stemness scores (RNAss) and DNA stemness scores (DNAss), which is measured the tumor stemness. We also discover an association between UCK2 expression and immune cells by CIBERSORT algorithm, ESTIMATE algorithm and ssGSEA algorithm, especially, related to T cell, monocytes, mast cells, and macrophages. This study aims to shed light on the role and possible mechanism of UCK2 in pan-cancer. Methods We used the R programming language for pan-cancer bulk sequencing data analysis, which were obtained from the University of California, Santa Cruz (UCSC) datasets. UCSC database is a very useful for explore data from TCGA and other cancer genomics datasets, The data we explored at the UCK2 transcriptome level came from TCGA data in the UCSC database. We explored differential UCK2 expression between tumor and normal samples. Immunohistochemistry (IHC) was utilized to validate the expression of UCK2 in different types cancers using tumor tissue chips. The correlations of UCK2 with prognosis, genetic instability, DNA repair, cancer stem cell characteristics, and immune cell infiltration were investigated. Furthermore, single-cell datasets, acquired from the Gene Expression Omnibus (GEO) database, were used to validate the relationship between UCK2 and immune cells. GEO is a famous public genomics database supporting freely disseminates microarray data. Finally, we analyzed the correlation between UCK2 and drug sensitivity. Results UCK2 expression was observed to be high in most cancers and was remarkably related to the prognosis of pan-cancers. We found that the increased UCK2 expression was associated with higher genetic instability. Additionally, positive relationships were observed between UCK2 expression and mismatch repair genes, homologous recombination repair genes, and cancer stemness across different cancer types. There were significant correlations between UCK2 and T cells, monocytes, mast cells, and macrophages. Moreover, as expected, the immune checkpoint human leucocyte antigen (HLA) was found to be negatively related to UCK2. Similarly, UCK2 was also observed to have a negative association with major histocompatibility complex (MHC) genes. We noted that UCK2 had significant correlations with the sensitivity to various anti-cancer drug. Conclusion We have observed that UCK2 plays pivotal roles in prognosis and tumor immunity, and it is associated with DNA repair and cancer stemness. The UCK2 gene exhibits a strong correlation with the immune checkpoints HLA. This study highlights its potential impact on drug sensitivity.
Collapse
Affiliation(s)
- Jinlong Tian
- Graduate School of Bengbu Medical University, Bengbu, Anhui, China
| | - Yanlei Li
- Graduate School of Bengbu Medical University, Bengbu, Anhui, China
| | - Yu Tong
- Sports Medicine Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yuan Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou, China
| | - Tingxiao Zhao
- Sports Medicine Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yao Kang
- Sports Medicine Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Qing Bi
- Graduate School of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
11
|
Jain S, Bhatt J, Gupta S, Bhatia DD. Nanotechnology at the crossroads of stem cell medicine. Biomater Sci 2024; 13:161-178. [PMID: 39584588 DOI: 10.1039/d4bm01257g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Nanotechnology in stem cell medicine is an interdisciplinary field which has gained a lot of interest recently. This domain addresses key challenges associated with stem cell medicine such as cell isolation, targeted delivery, and tracking. Nanotechnology-based approaches, including magnetic cell sorting, fluorescent tagging, and drug or biomolecule conjugation for delivery, have enhanced precision in stem cell isolation and guided cell migration, increasing the therapeutic potential. Recent studies have focused on using nanomaterials and scaffolds to drive stem cell differentiation by activating specific molecular pathways, achieved through embedding biomolecules within the scaffold or through the scaffold's material composition and structure alone. These innovations hold promise in therapeutic applications across various diseases, including cancer stem cell targeting, neurodegenerative disorders, pre-eclampsia, cardiovascular conditions, and organoid development. This review examines recent advancements in the field, explores potential applications like biosensors and nanochips, and highlights the challenges and research gaps.
Collapse
Affiliation(s)
- Sweny Jain
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Jay Bhatt
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Sharad Gupta
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| | - Dhiraj Devidas Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Palaj, Gujarat, 382355, India.
| |
Collapse
|
12
|
Qin H, Zhang Q, Guo Y. Genome-wide identification of alternative splicing related with transcription factors and splicing regulators in breast cancer stem cells responding to fasting-mimicking diet. Comput Biol Chem 2024; 113:108272. [PMID: 39509796 DOI: 10.1016/j.compbiolchem.2024.108272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Fasting-mimicking diet (FMD) can effectively inhibit the viability of breast cancer stem cells (CSCs). However, the molecular mechanisms underlying the inhibitory function of FMD on breast CSCs remain largely unknown. Elucidating the mechanisms by which FMD suppresses breast CSCs is beneficial to targeting breast CSCs. Herein, we systematically analyze alternative splicing and RNA binding protein (RBP) expression in breast CSCs during FMD. The analysis results show that a large number of regulated alternative splicing (RAS) and differentially expressed genes (DEGs) appear responding to FMD. Further studies show that there are potential regulatory relationships between transcription factors (TFs) with RAS (RAS-TFs) and their differentially expressed target genes (RAS-TF-DEGs). Moreover, differentially expressed RNA binding proteins (DERBPs) exhibit potential regulatory functions on RAS-TFs. In short, DERBPs potentially control the alternative splicing of TFs (RAS-TFs), regulating their target gene (RAS-TF-DEG) expression, which leads to the regulation of biological processes in breast CSCs during FMD. In addition, the alternative splicing and DEGs are compared between breast CSCs and differentiated cancer cells during FMD, providing new interpretations for the different responses of the two types of cells. Our studies will shed light on the understanding of the molecular mechanisms underlying breast CSC inhibition induced by FMD.
Collapse
Affiliation(s)
- Hongshuang Qin
- Department of Biological and Food Engineering, Lyuliang University, Lvliang, Shanxi 033001, China.
| | - Qian Zhang
- Department of Biological and Food Engineering, Lyuliang University, Lvliang, Shanxi 033001, China
| | - Yanxiang Guo
- Department of Biological and Food Engineering, Lyuliang University, Lvliang, Shanxi 033001, China
| |
Collapse
|
13
|
Wang D, Duan JJ, Guo YF, Chen JJ, Chen TQ, Wang J, Yu SC. Targeting the glutamine-arginine-proline metabolism axis in cancer. J Enzyme Inhib Med Chem 2024; 39:2367129. [PMID: 39051546 PMCID: PMC11275534 DOI: 10.1080/14756366.2024.2367129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 04/27/2024] [Accepted: 06/06/2024] [Indexed: 07/27/2024] Open
Abstract
Metabolic abnormalities are an important feature of tumours. The glutamine-arginine-proline axis is an important node of cancer metabolism and plays a major role in amino acid metabolism. This axis also acts as a scaffold for the synthesis of other nonessential amino acids and essential metabolites. In this paper, we briefly review (1) the glutamine addiction exhibited by tumour cells with accelerated glutamine transport and metabolism; (2) the methods regulating extracellular glutamine entry, intracellular glutamine synthesis and the fate of intracellular glutamine; (3) the glutamine, proline and arginine metabolic pathways and their interaction; and (4) the research progress in tumour therapy targeting the glutamine-arginine-proline metabolic system, with a focus on summarising the therapeutic research progress of strategies targeting of one of the key enzymes of this metabolic system, P5CS (ALDH18A1). This review provides a new basis for treatments targeting the metabolic characteristics of tumours.
Collapse
Affiliation(s)
- Di Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
| | - Jiang-jie Duan
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
- Jin-feng Laboratory, Chongqing, China
| | - Yu-feng Guo
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jun-jie Chen
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
| | - Tian-qing Chen
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
- Jin-feng Laboratory, Chongqing, China
| | - Shi-cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
- Jin-feng Laboratory, Chongqing, China
| |
Collapse
|
14
|
Zhu H, Xie Z. Therapeutic potential of tLyp-1-EV-shCTCF in inhibiting liver cancer stem cell self-renewal and immune escape via SALL3 modulation in hepatocellular carcinoma. Transl Oncol 2024; 49:102048. [PMID: 39186862 PMCID: PMC11388803 DOI: 10.1016/j.tranon.2024.102048] [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: 11/09/2023] [Revised: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 08/28/2024] Open
Abstract
The progression of hepatocellular carcinoma (HCC) is influenced by disrupted metabolic processes, presenting challenges in prognostic outcomes. Hepatocellular carcinoma (HCC), a leading cause of cancer-related mortality, is closely associated with metabolic reprogramming and stem cell-like properties in liver cancer stem cells (LCSCs). This study explored the potential molecular mechanisms by which tLyP-1-modified extracellular vesicles (EVs) delivering CTCF shRNA (tLyp-1-EV-shCTCF) regulate mitochondrial DNA methylation-induced glycolytic metabolic reprogramming and LCSC self-renewal. Through a series of methods, including Western blot, nanoparticle tracking analysis, and immunofluorescence, we demonstrated the successful delivery and internalization of tLyp-1-EV in HCC cells. Our results identified SALL3 as a critical factor underexpressed in HCC and LCSCs, while CTCF was overexpressed. Overexpression of SALL3 inhibited LCSC self-renewal and immune evasion by blocking the CTCF-DNMT3A interaction, thus repressing DNMT3A methyltransferase activity and subsequent mitochondrial DNA methylation-mediated glycolytic metabolic reprogramming. In vivo experiments further supported these findings, showing that tLyp-1-EV-shCTCF treatment significantly reduced tumor growth by upregulating SALL3 expression, thereby inhibiting glycolytic metabolic reprogramming and enhancing the immune response against HCC cells. This study provides novel insights into the role of SALL3 and mitochondrial DNA methylation in HCC progression, offering potential therapeutic targets for combating HCC and its stem cell-like properties.
Collapse
Affiliation(s)
- Heng Zhu
- Department of Gastroenterology, The Fourth People's Hospital of Jinan, No.50, Normal Road, Tianqiao District, Jinan, Shandong Province 250031, P R China.
| | - Zhihui Xie
- Department of infectious diseases, Zibo Central Hospital, Zibo 255000, P R China
| |
Collapse
|
15
|
Sato T, Oshi M, Huang JL, Chida K, Roy AM, Endo I, Takabe K. CD133 expression is associated with less DNA repair, better response to chemotherapy and survival in ER-positive/HER2-negative breast cancer. Breast Cancer Res Treat 2024; 208:415-427. [PMID: 39017815 PMCID: PMC11849687 DOI: 10.1007/s10549-024-07434-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE CD133, a cancer stem cells (CSC) marker, has been reported to be associated with treatment resistance and worse survival in triple-negative breast cancer (BC). However, the clinical relevance of CD133 expression in ER-positive/HER2-negative (ER + /HER2-) BC, the most abundant subtype, remains unknown. METHODS The BC cohorts from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC, n = 1904) and The Cancer Genome Atlas (TCGA, n = 1065) were used to obtain biological variables and gene expression data. RESULTS Epithelial cells were the exclusive source of CD133 gene expression in a bulk BC. CD133-high ER + /HER2- BC was associated with CD24, NOTCH1, DLL1, and ALDH1A1 gene expressions, as well as with WNT/β-Catenin, Hedgehog, and Notch signaling pathways, all characteristic for CSC. Consistent with a CSC phenotype, CD133-low BC was enriched with gene sets related to cell proliferation, such as G2M Checkpoint, MYC Targets V1, E2F Targets, and Ki67 gene expression. CD133-low BC was also linked with enrichment of genes related to DNA repair, such as BRCA1, E2F1, E2F4, CDK1/2. On the other hand, CD133-high tumors had proinflammatory microenvironment, higher activity of immune cells, and higher expression of genes related to inflammation and immune response. Finally, CD133-high tumors had better pathological complete response after neoadjuvant chemotherapy in GSE25066 cohort and better disease-free survival and overall survival in both TCGA and METABRIC cohorts. CONCLUSION CD133-high ER + /HER2- BC was associated with CSC phenotype such as less cell proliferation and DNA repair, but also with enhanced inflammation, better response to neoadjuvant chemotherapy and better prognosis.
Collapse
Affiliation(s)
- Takumi Sato
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- University of Tokyo Hospital, Tokyo, 113-8655, Japan
- National Hospital Organization Disaster Medical Center, Tokyo, 190-0014, Japan
| | - Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Jing Li Huang
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
| | - Kohei Chida
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Arya Mariam Roy
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA.
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, 14263, USA.
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8520, Japan.
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan.
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402, Japan.
| |
Collapse
|
16
|
Ali LS, Attia YAM, Mourad S, Halawa EM, Abd Elghaffar NH, Shokry S, Attia OM, Makram M, Wadan AHS, Negm WA, Elekhnawy E. The missing link between cancer stem cells and immunotherapy. Curr Med Res Opin 2024; 40:1963-1984. [PMID: 39316769 DOI: 10.1080/03007995.2024.2407963] [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: 06/17/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Cancer stem cells (CSCs) are cancer cells that can self-renew and give rise to tumors. The multipotency of CSCs enables the generation of diverse cancer cell types and their potential for differentiation and resilience against chemotherapy and radiation. Additionally, specific biomarkers have been identified for them, such as CD24, CD34, CD44, CD47, CD90, and CD133. The CSC model suggests that a subset of CSCs within tumors is responsible for tumor growth. The tumor microenvironment (TME), including fibroblasts, immune cells, adipocytes, endothelial cells, neuroendocrine (NE) cells, extracellular matrix (ECM), and extracellular vesicles, has a part in shielding CSCs from the host immune response as well as protecting them against anticancer drugs. The regulation of cancer stem cell plasticity by cancer-associated fibroblasts (CAFs) occurs through specific signaling pathways that differ among various types of cancer, utilizing the IGF-II/IGF1R, FAK, and c-Met/FRA1/HEY1 signaling pathways. Due to the intricate dynamics of CSC proliferation, controlling their growth necessitates innovative approaches and much more research. Our current review speculates an outline of how the TME safeguards stem cells, their interaction with CSCs, and the involvement of the immune and inflammatory systems in CSC differentiation and maintenance. Several technologies have the ability to identify CSCs; however, each approach has limitations. We discuss how these methods can aid in recognizing CSCs in several cancer types, comprising brain, breast, liver, stomach, and colon cancer. Furthermore, we explore different immunotherapeutic strategies targeting CSCs, including stimulating cancer-specific T cells, modifying immunosuppressive TMEs, and antibody-mediated therapy targeting CSC markers.
Collapse
Affiliation(s)
- Lobna Safwat Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | | | - Sohaila Mourad
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Esraa M Halawa
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Seham Shokry
- Faculty of Science, Tanta University, Tanta, Egypt
| | - Omar M Attia
- Faculty of Medicine, Cairo University, Giza, Egypt
| | - Maha Makram
- Faculty of Science, Zagazig University, Zagazig, Egypt
| | | | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
17
|
Li B, Tian J, Zhang F, Wu C, Li Z, Wang D, Zhuang J, Chen S, Song W, Tang Y, Ping Y, Liu B. Self-assembled aldehyde dehydrogenase-activatable nano-prodrug for cancer stem cell-enriched tumor detection and treatment. Nat Commun 2024; 15:9417. [PMID: 39482286 PMCID: PMC11528051 DOI: 10.1038/s41467-024-53771-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
Cancer stem cells, characterized by high tumorigenicity and drug-resistance, are often responsible for tumor progression and metastasis. Aldehyde dehydrogenases, often overexpressed in cancer stem cells enriched tumors, present a potential target for specific anti-cancer stem cells treatment. In this study, we report a self-assembled nano-prodrug composed of aldehyde dehydrogenases activatable photosensitizer and disulfide-linked all-trans retinoic acid for diagnosis and targeted treatment of cancer stem cells enriched tumors. The disulfide-linked all-trans retinoic acid can load with photosensitizer and self-assemble into a stable nano-prodrug, which can be disassembled into all-trans retinoic acid and photosensitizer in cancer stem cells by high level of glutathione. As for the released photosensitizer, overexpressed aldehyde dehydrogenase catalyzes the oxidation of aldehydes to carboxyl under cancer stem cells enriched microenvironment, activating the generation of reactive oxygen species and fluorescence emission. This generation of reactive oxygen species leads to direct killing of cancer stem cells and is accompanied by a noticeable fluorescence enhancement for real-time monitoring of the cancer stem cells enriched microenvironment. Moreover, the released all-trans retinoic acid, as a differentiation agent, reduce the cancer stem cells stemness and improve the cancer stem cells enriched microenvironment, offering a synergistic effect for enhanced anti-cancer stem cells treatment of photosensitizer in inhibition of in vivo tumor growth and metastasis.
Collapse
Affiliation(s)
- Bowen Li
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Jianwu Tian
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Fu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chongzhi Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhiyao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dandan Wang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Jiahao Zhuang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Siqin Chen
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Wentao Song
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yufu Tang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.
| |
Collapse
|
18
|
Nie X, Liu J, Wang D, Li C, Teng Y, Li Z, Jia Y, Wang P, Deng J, Li W, Lu L. MiR-21-5p Modulates Cisplatin-Resistance of CD44+ Gastric Cancer Stem Cells Through Regulating the TGF-β2/SMAD Signaling Pathway. Int J Gen Med 2024; 17:4579-4593. [PMID: 39411053 PMCID: PMC11476341 DOI: 10.2147/ijgm.s476647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Background Cisplatin (DDP) resistance in gastric cancer (GC) is likely to come from gastric cancer stem cells (GCSC). It is a new idea to study the mechanism of the DDP-resistance in GCSC from miRNA. Materials and Methods CD44+ GCSCs and CD44- control cells were constructed based on the HGC27 gastric cancer cell line. DDP sensitivities in CD44+ and CD44- cells were detected via CCK-8 assay. The differential expression of miR-21-5p in these cell lines was detected by RT‒qPCR. The expression levels of downstream TGF-β2, SMAD2 and SMAD3 were determined through RT‒PCR and Western blotting. A luciferase assay was used to detect the relationship between miR-21-5p and TGFB2, and the TCGA database, clinical data from our centre, and vivo experiment were used for validation. Finally, we knocked down miR-21-5p to detect changes in cisplatin resistance in GCSCs and to verify changes in the levels of downstream pathways in GCSCs. Results CD44+ GCSCs induced cisplatin resistance compared with CD44- cells. miR-21-5p was highly expressed in GCSCs, and the TGF-β2/SMAD pathway was also highly expressed. TGFB2 was proven to be a downstream target gene of miR-21-5p and had a positive relationship with it in phenotype. After knockdown of miR-21-5p, the TGF-β2/SMAD pathway was also inhibited, and the resistance of GCSCs to cisplatin was specifically decreased. Conclusion MiR-21-5p promotes cisplatin resistance in gastric cancer stem cells by regulating the TGF-β2/SMAD signalling pathway.
Collapse
Affiliation(s)
- Xinyang Nie
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060, People’s Republic of China
| | - Jian Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Daohan Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Chuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yuxin Teng
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Zhufeng Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yangpu Jia
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Peiyao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Jingyu Deng
- Tianjin Medical University, Tianjin, People’s Republic of China
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060, People’s Republic of China
| | - Weidong Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| | - Li Lu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
- Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
19
|
Zhang X, Du W, Huang X, Zhong H, Hu N. An overview of current research on cancer stem cells: a bibliometric analysis. Clin Transl Oncol 2024; 26:2466-2478. [PMID: 38625493 DOI: 10.1007/s12094-024-03486-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Cancer stem cells (CSCs) represent a potential mechanism contributing to tumorigenesis, metastasis, recurrence, and drug resistance. The objective of this study is to investigate the status quo and advancements in CSC research utilizing bibliometric analysis. METHODS Publications related to CSCs from 2010 to 2022 were collected from the Web of Science Core Collection database. Various analytical tools including CiteSpace, VOSviewer, Scimago Graphica, and GraphPad Prism were used to visualize aspects such as co-authorship, co-occurrence, and co-citation within CSC research to provide an objective depiction of the contemporary status and developmental trajectory of the CSC field. RESULTS A total of 22,116 publications were included from 1942 journals written by 95,992 authors. Notably, China emerged as the country with the highest number of publications, whereas the United States exerted the most significant influence within the field. MD Anderson Cancer Center emerged as the institution making the most comprehensive contributions. Wicha M.S. emerged as the most prolific and influential researcher. Among journals, Cancers emerged as a focal point for CSC research, consistently publishing a wealth of high-quality papers. Furthermore, it was observed that most journals tended to approach CSC research from molecular, biological, and immunological perspectives. The research into CSCs encompassed a broad array of topics, including isolation and enrichment techniques, biomarkers, biological characteristics, cancer therapy strategies, and underlying biological regulatory mechanisms. Notably, exploration of the tumor microenvironment and extracellular vesicles emerged as burgeoning research frontiers for CSCs. CONCLUSION The research on CSCs has garnered growing interest. A trend toward multidisciplinary homogeneity is emerging within the realm of CSCs. Further investigation could potentially center on the patients of extracellular vesicles and the tumor microenvironment in relation to CSCs.
Collapse
Affiliation(s)
- Xueyang Zhang
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Wenbo Du
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Xizhi Huang
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Haoting Zhong
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Ning Hu
- The First Affiliated Hospital, Chongqing Medical University, No. 1 of Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
20
|
Kong W, Gao Y, Zhao S, Yang H. Cancer stem cells: advances in the glucose, lipid and amino acid metabolism. Mol Cell Biochem 2024; 479:2545-2563. [PMID: 37882986 DOI: 10.1007/s11010-023-04861-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/13/2023] [Indexed: 10/27/2023]
Abstract
Cancer stem cells (CSCs) are a class of cells with self-renewal and multi-directional differentiation potential, which are present in most tumors, particularly in aggressive tumors, and perform a pivotal role in recurrence and metastasis and are expected to be one of the important targets for tumor therapy. Studies of tumor metabolism in recent years have found that the metabolic characteristics of CSCs are distinct from those of differentiated tumor cells, which are unique to CSCs and contribute to the maintenance of the stemness characteristics of CSCs. Moreover, these altered metabolic profiles can drive the transformation between CSCs and non-CSCs, implying that these metabolic alterations are important markers for CSCs to play their biological roles. The identification of metabolic changes in CSCs and their metabolic plasticity mechanisms may provide some new opportunities for tumor therapy. In this paper, we review the metabolism-related mechanisms of CSCs in order to provide a theoretical basis for their potential application in tumor therapy.
Collapse
Affiliation(s)
- Weina Kong
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China
| | - Yunge Gao
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China
| | - Shuhua Zhao
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China
| | - Hong Yang
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China.
| |
Collapse
|
21
|
Tan Z, Pan K, Sun M, Pan X, Yang Z, Chang Z, Yang X, Zhu J, Zhan L, Liu Y, Li X, Lin K, Chen L, Mo H, Luo W, Kan C, Duan L, Zheng H. CCKBR+ cancer cells contribute to the intratumor heterogeneity of gastric cancer and confer sensitivity to FOXO inhibition. Cell Death Differ 2024; 31:1302-1317. [PMID: 39164456 PMCID: PMC11445462 DOI: 10.1038/s41418-024-01360-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: 01/11/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
The existence of heterogeneity has plunged cancer treatment into a challenging dilemma. We profiled malignant epithelial cells from 5 gastric adenocarcinoma patients through single-cell sequencing (scRNA-seq) analysis, demonstrating the heterogeneity of gastric adenocarcinoma (GA), and identified the CCKBR+ stem cell-like cancer cells associated poorly differentiated and worse prognosis. We further conducted targeted analysis using single-cell transcriptome libraries, including 40 samples, to confirm these screening results. In addition, we revealed that FOXOs are involved in the progression and development of CCKBR+ gastric adenocarcinoma. Inhibited the expression of FOXOs and disrupting cancer cell stemness reduce the CCKBR+ GA organoid formation and impede tumor progression. Mechanically, CUT&Tag sequencing and Lectin pulldown revealed that FOXOs can activate ST3GAL3/4/5 as well as ST6GALNAC6, promoting elevated sialyation levels in CCKBR+ tumor cells. This FOXO-sialyltransferase axis contributes to the maintenance of homeostasis and the growth of CCKBR+ tumor cells. This insight provides novel perspectives for developing targeted therapeutic strategies aimed at the treating CCKBR associated gastric cancer.
Collapse
Affiliation(s)
- Zhenya Tan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ke Pan
- Department of General Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Minqiong Sun
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xianzhu Pan
- Department of Pathology and Pathophysiology, School of Basic Medicine, Anhui Medical College, Hefei, 230032, China
| | - Zhi Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zhiling Chang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xue Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jicheng Zhu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Li Zhan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yakun Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xiaofei Li
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Keqiong Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Lin Chen
- Department of General Surgery, Anhui Provincial Cancer Hospital, Hefei, 230032, China
| | - Hui Mo
- Department of General Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wei Luo
- Department of General Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chen Kan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Lunxi Duan
- Department of General Surgery, the Second Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Hong Zheng
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
22
|
Kim JH, Lee J, Lee KW, Xiong H, Li M, Kim JS. Trapped in Cells: A Selective Accumulation Approach for Type-I Photodynamic Ablation of Cancer Stem-like Cells. JACS AU 2024; 4:3657-3667. [PMID: 39328753 PMCID: PMC11423316 DOI: 10.1021/jacsau.4c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024]
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme responsible for converting aldehyde functional groups into carboxylate metabolites. Elevated ALDH activity is a characteristic feature of cancer stem-like cells (CSCs). As a novel approach to target the CSC trait of overexpressing ALDH, we aimed to utilize ALDH activity for the selective accumulation of a photosensitizer in ALDHHigh CSCs. A novel ALDH substrate photosensitizer, SCHO, with thionylated coumarin and N-ethyl-4-(aminomethyl)benzaldehyde was developed to achieve this goal. Our study demonstrated the efficient metabolism of the aldehyde unit of SCHO into carboxylate, leading to its accumulation in ALDHHigh MDA-MB-231 cells. Importantly, we established the selectivity of SCHO as an ALDHHigh cell photosensitizer as it is not a substrate for ABC transporters. SCHO-based photodynamic therapy triggers apoptosis and pyroptosis in MDA-MB-231 cells and further reduces the characteristics of CSCs. Our study presents a novel strategy to target CSCs by exploiting their cellular metabolism to enhance photosensitizer accumulation, highlighting the potential of photodynamic therapy as a powerful tool for eliminating ALDHHigh CSCs.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- Department
of Chemical and Systems Biology, Chem-H
and Stanford Cancer Institute, Stanford School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Jieun Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Kyung-Woo Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Hao Xiong
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
23
|
Razavi Z, Soltani M, Pazoki-Toroudi H, Dabagh M. Microfluidic systems for modeling digestive cancer: a review of recent progress. Biomed Phys Eng Express 2024; 10:052002. [PMID: 39142294 DOI: 10.1088/2057-1976/ad6f15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/14/2024] [Indexed: 08/16/2024]
Abstract
Purpose. This review aims to highlight current improvements in microfluidic devices designed for digestive cancer simulation. The review emphasizes the use of multicellular 3D tissue engineering models to understand the complicated biology of the tumor microenvironment (TME) and cancer progression. The purpose is to develop oncology research and improve digestive cancer patients' lives.Methods. This review analyzes recent research on microfluidic devices for mimicking digestive cancer. It uses tissue-engineered microfluidic devices, notably organs on a chip (OOC), to simulate human organ function in the lab. Cell cultivation on modern three-dimensional hydrogel platforms allows precise geometry, biological components, and physiological qualities. The review analyzes novel methodologies, key findings, and technical progress to explain this field's advances.Results. This study discusses current advances in microfluidic devices for mimicking digestive cancer. Micro physiological systems with multicellular 3D tissue engineering models are emphasized. These systems capture complex biochemical gradients, niche variables, and dynamic cell-cell interactions in the tumor microenvironment (TME). These models reveal stomach cancer biology and progression by duplicating the TME. Recent discoveries and technology advances have improved our understanding of gut cancer biology, as shown in the review.Conclusion. Microfluidic systems play a crucial role in modeling digestive cancer and furthering oncology research. These platforms could transform drug development and treatment by revealing the complex biology of the tumor microenvironment and cancer progression. The review provides a complete summary of recent advances and suggests future research for field professionals. The review's major goal is to further medical research and improve digestive cancer patients' lives.
Collapse
Affiliation(s)
- ZahraSadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran
- Biochemistry Research Center, Iran University Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K N Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
- Centre for Sustainable Business, International Business University, Toronto, Canada
| | | | - Mahsa Dabagh
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, WI 53211, United States of America
| |
Collapse
|
24
|
Obaid Saleh R, Shbeer AM, Jetti R, Ahmed Robadi I, Hjazi A, Hussein Kareem A, Noori Shakir M, Qasim Alasheqi M, Alawadi A, Haslany A. Association between lncRNAs with stem cells in cancer; a particular focus on lncRNA-CSCs axis in cancer immunopathogenesis. Int Immunopharmacol 2024; 136:112306. [PMID: 38833843 DOI: 10.1016/j.intimp.2024.112306] [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: 04/02/2024] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024]
Abstract
A unique population of cells known as cancer stem cells (CSCs) is essential to developing and spreading cancer. Cancer initiation, maintenance, and progression are all believed to be significantly impacted by the distinct characteristics these cells exhibit regarding self-renewal, proliferation, and differentiation. Transcriptional, post-transcriptional, and translational processes are the only steps of gene expression that lncRNAs can affect. As a result, these proteins participate in numerous biological processes, including the repair of DNA damage, inflammatory reactions, metabolic control, the survival of cells, intercellular communication, and the development and specialization of cells. Studies have indicated that lncRNAs are important for controlling the increase in the subset of CSCs contributing to cancer development. The knowledge that is currently available about lncRNAs and their critical role in maintaining the biological properties of CSCs is highlighted in this study.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Abdullah M Shbeer
- Department of Surgery, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia.
| | - Raghu Jetti
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ibrahim Ahmed Robadi
- Department of Pathology, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | - Ahmed Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | - Ali Haslany
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| |
Collapse
|
25
|
Zhang J, Cui T, Xu J, Wang P, Lv C, Pan G. The potential of cancer stem cells for personalized risk assessment and therapeutic intervention in individuals with intrahepatic cholangiocarcinoma. Discov Oncol 2024; 15:306. [PMID: 39048806 PMCID: PMC11269542 DOI: 10.1007/s12672-024-01179-7] [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: 04/24/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Accumulating evidence suggests that intrahepatic cholangiocarcinoma (ICC) is a stem cell-based disease, but information on the biology of cancer stem cells (CSC) in ICC is very limited. METHODS ICC RNA-seq cohorts from three different public databases were integrated and the protein-coding genes were divided into different modules using "WGCNA" to screen the most relevant modules with CSC scores. Least Absolute Shrinkage and Selection Operator (LASSO) regression were introduced to construct prognostic classification models. In addition, the extent of immune cell infiltration in patients in different risk groups was assessed based on the ESTIMATE, CIBERSORT, MCP-Counter, and single sample gene set enrichment analysis (ssGSEA) algorithms. Finally, the correlation between different risk scores and common drugs was analyzed by pRRophetic package and Spearman method. RESULTS In the present study, we found that a high CSC score was associated with a poorer prognosis in patients with ICC. The yellow module obtained by WGCNA was significantly positively correlated with the CSCs score, in which 8 genes were served to build a prognostic classification model, and the obtained risk score was negatively correlated with CSCs score and prognosis. The low-risk score was more suitable for immunotherapy, and the high-risk score was more suitable for treatment with 11 antitumor drugs. CONCLUSION This study revealed the regulatory role of CSC-mediated EMT, angiogenesis, and immunomodulatory biological processes in ICC, and applied a prognostic classification model to highlight the great potential of CSC for personalized risk assessment, chemotherapy, and immunotherapy intervention in ICC individuals.
Collapse
Affiliation(s)
- Jian Zhang
- Hepatobiliary Surgery, Shengli Oilfield Central Hospital, Dongying, 257093, China
| | - Tao Cui
- Hepatobiliary Surgery, Shengli Oilfield Central Hospital, Dongying, 257093, China
| | - Jiaobang Xu
- Hepatobiliary Surgery, Shengli Oilfield Central Hospital, Dongying, 257093, China
| | - Peng Wang
- Hepatobiliary Surgery, Shengli Oilfield Central Hospital, Dongying, 257093, China
| | - Chongqing Lv
- Hepatobiliary Surgery, Shengli Oilfield Central Hospital, Dongying, 257093, China
| | - Guozheng Pan
- Hepatobiliary Surgery, Shengli Oilfield Central Hospital, Dongying, 257093, China.
| |
Collapse
|
26
|
Miki K, Oe M, Suzuki K, Miki K, Mu H, Kato Y, Iwatake M, Yukawa H, Baba Y, Ueda Y, Mori Y, Ohe K. Dual-responsive near-infrared turn-on fluorescent probe for cancer stem cell-specific visualization. J Mater Chem B 2024; 12:6959-6967. [PMID: 38913327 DOI: 10.1039/d4tb00897a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Aldehyde dehydrogenase 1A1 (ALDH1A1) stands out as one of the most reliable intracellular biomarkers for stem cells because it is expressed in both cancer stem cells (CSCs) and normal somatic stem cells (NSCs). Although several turn-on fluorescent probes for ALDH1A1 have been developed to visualize CSCs in cancer cells, the discrimination of CSCs from NSCs is difficult. We here report an AND-type dual-responsive fluorescent probe, CHO_βgal, the near-infrared fluorescence of which can be turned on after responding to both ALDH1A1 and β-galactosidase. The AND-type dual responsiveness enables CSCs to be clearly visualized, whereas NSCs are non-emissive in microscopy. CSC-positive metastasis model lungs were successfully discriminated from normal lungs in ex vivo staining experiments using CHO_βgal, whereas the single-input ALDH1A1-responsive probe failed to achieve this discrimination owing to pronounced false-positive fluorescence output from lung NSCs. In tissue slice staining experiments, even in the presence of adjacent normal tissues, the peripheral region-specific localization of CSCs was clear. The versatility of CHO_βgal holds promise not only as a fundamental in vitro research tool for visualizing CSCs but also as a valuable asset in practical tissue staining diagnosis, significantly contributing to the assessment of cancer malignancy.
Collapse
Affiliation(s)
- Koji Miki
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Masahiro Oe
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Kanae Suzuki
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Koki Miki
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Huiying Mu
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Yoshimi Kato
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Mayumi Iwatake
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Hiroshi Yukawa
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
- Department of Quantum Life Science, Graduate School of Science, Chiba University, Chiba 265-8522, Japan
| | - Yoshinobu Baba
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Yoshifumi Ueda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kouichi Ohe
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| |
Collapse
|
27
|
Zhang L, Hu X, Xiao K, Kong L. Effective identification and differential analysis of anticancer peptides. Biosystems 2024; 241:105246. [PMID: 38848816 DOI: 10.1016/j.biosystems.2024.105246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Anticancer peptides (ACPs) have recently emerged as promising cancer therapeutics due to their selectivity and lower toxicity. However, the number of experimentally validated ACPs is limited, and identifying ACPs from large-scale sequence data is time-consuming and expensive. Therefore, it is critical to develop and improve upon existing computational models for identifying ACPs. In this study, a computational method named ACP_DA was proposed based on peptide residue composition and physiochemical properties information. To curtail overfitting and reduce computational costs, a sequential forward selection method was utilized to construct the optimal feature groups. Subsequently, the feature vectors were fed into light gradient boosting machine classifier for model construction. It was observed by an independent set test that ACP_DA achieved the highest Matthew's correlation coefficient of 0.63 and accuracy of 0.8129, displaying at least a 2% enhancement compared to state-of-the-art methods. The satisfactory results demonstrate the effectiveness of ACP_DA as a powerful tool for identifying ACPs, with the potential to significantly contribute to the development and optimization of promising therapies. The data and resource codes are available at https://github.com/Zlclab/ACP_DA.
Collapse
Affiliation(s)
- Lichao Zhang
- School of Mathematics and Statistics, Northeastern University at Qinhuangdao, Qinhuangdao, PR China; Hebei Innovation Center for Smart Perception and Applied Technology of Agricultural Data, Qinhuangdao, PR China
| | - Xueli Hu
- School of Mathematics and Statistics, Northeastern University at Qinhuangdao, Qinhuangdao, PR China
| | - Kang Xiao
- School of Mathematics and Statistics, Northeastern University at Qinhuangdao, Qinhuangdao, PR China
| | - Liang Kong
- Hebei Innovation Center for Smart Perception and Applied Technology of Agricultural Data, Qinhuangdao, PR China; School of Mathematics and Information Science & Technology, Hebei Normal University of Science & Technology, Qinhuangdao, PR China.
| |
Collapse
|
28
|
Li X, Zhang C, Yue W, Jiang Y. Modulatory effects of cancer stem cell-derived extracellular vesicles on the tumor immune microenvironment. Front Immunol 2024; 15:1362120. [PMID: 38962016 PMCID: PMC11219812 DOI: 10.3389/fimmu.2024.1362120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Cancer stem cells (CSCs), accounting for only a minor cell proportion (< 1%) within tumors, have profound implications in tumor initiation, metastasis, recurrence, and treatment resistance due to their inherent ability of self-renewal, multi-lineage differentiation, and tumor-initiating potential. In recent years, accumulating studies indicate that CSCs and tumor immune microenvironment act reciprocally in driving tumor progression and diminishing the efficacy of cancer therapies. Extracellular vesicles (EVs), pivotal mediators of intercellular communications, build indispensable biological connections between CSCs and immune cells. By transferring bioactive molecules, including proteins, nucleic acids, and lipids, EVs can exert mutual influence on both CSCs and immune cells. This interaction plays a significant role in reshaping the tumor immune microenvironment, creating conditions favorable for the sustenance and propagation of CSCs. Deciphering the intricate interplay between CSCs and immune cells would provide valuable insights into the mechanisms of CSCs being more susceptible to immune escape. This review will highlight the EV-mediated communications between CSCs and each immune cell lineage in the tumor microenvironment and explore potential therapeutic opportunities.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Animal Science, College of Animal Science, Hebei North University, Zhangjiakou, Hebei, China
- Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Cuilian Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Wei Yue
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
| | - Yuening Jiang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
| |
Collapse
|
29
|
Liu N, Zheng Q, Zhang Y, Wang H, Zhang Z, He L, Wei C, Xia H, Liu Y, Wang X. Hypoxia differently regulates the proportion of ALDH hi cells in lung squamous carcinoma H520 and adenocarcinoma A549 cells via the Wnt/β-catenin pathway. Thorac Cancer 2024; 15:1419-1428. [PMID: 38736300 PMCID: PMC11194122 DOI: 10.1111/1759-7714.15328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a specific subpopulation of cancer cells with the ability of self-renewal, infinite proliferation, multidifferentiation and tumorigenicity, and play critical roles in cancer progression and treatment resistance. CSCs are tightly regulated by the tumor microenvironment, such as hypoxia; however, how hypoxia regulates CSCs in non-small cell lung cancer (NSCLC) remains unclear. METHODS The proportion of ALDHhi cells was examined using the Aldefluor assay. Tankyrase inhibitor XAV939 and siRNA were used to inhibit β-catenin while pcDNA3-β-catenin (S33Y) plasmid enhanced the expression of β-catenin. Western blot was administered for protein detection. The mRNA expression was measured by quantitative real-time PCR. RESULTS We found that hypoxia led to an increase in the proportion of ALDHhi cells in lung squamous carcinoma (LUSC) H520 cells, while causing a decrease in the ALDHhi cell proportion in lung adenocarcinoma (LUAD) A549 cells. Similarly, β-catenin expression was upregulated in H520 cells but downregulated in A549 cells upon exposure to hypoxia. Mechanically, the proportion of ALDHhi cells in both cell lines was decreased by β-catenin inhibitor or siRNA knockdown, whereas increased after β-catenin overexpression. Furthermore, hypoxia treatment suppressed E-cadherin expression in H520 cells and enhanced N-cadherin and β-catenin expression, while this effect was completely opposite in A549 cells. CONCLUSION The hypoxia-EMT-β-catenin axis functions as an important regulator for the proportion of CSCs in NSCLC and could potentially be explored as therapeutic targets in the future.
Collapse
Affiliation(s)
- Ni Liu
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Qi Zheng
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Yuqing Zhang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Huimin Wang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
- Department of OncologyYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Zhihui Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Long He
- Department of OncologyThe Third Hospital of JinanJinanChina
| | - Chenxi Wei
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Handai Xia
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Yanguo Liu
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| | - Xiuwen Wang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
30
|
Chen C, Huang R, Wang N, Zheng Y, Zhou J, Yang B, Wang X, Zhang J, Pan B, Chen Z, Wang S, Wang Z, Xiang S. Fu-Zheng-Yi-Liu Formula inhibits the stem cells and metastasis of prostate cancer via tumor-associated macrophages/C-C motif chemokine ligand 5 pathway in tumor microenvironment. Chin J Nat Med 2024; 22:501-514. [PMID: 38906598 DOI: 10.1016/s1875-5364(24)60653-9] [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: 12/12/2023] [Indexed: 06/23/2024]
Abstract
Prostate cancer (PCa) is the second most common malignancy among men globally. The Fu-Zheng-Yi-Liu (FZYL) Formula has been widely utilized in the treatment of PCa. This study investigates whether the FZYL Formula can inhibit PCa by targeting the TAMs/CCL5 pathway. We conducted in vitro co-cultures and in vivo co-injections of PCa cells and TAMs to mimic their interaction. Results showed that the FZYL Formula significantly reduced the proliferation, colony formation, subpopulations of PCSCs, and sphere-formation efficacy of PCa cells, even in the presence of TAM co-culture. Additionally, the Formula markedly decreased the migration, invasion, and epithelial-mesenchymal transition (EMT) of PCa cells induced by TAMs. The FZYL Formula also reversed M2 phenotype polarization in TAMs and dose-dependently reduced their CCL5 expression and secretion, with minimal cytotoxicity observed. Mechanistic studies confirmed that the TAMs/CCL5 axis is a critical target of the FZYL Formula, as the addition of exogenous CCL5 partially reversed the formula's inhibitory effects on PCSCs self-renewal in the co-culture system. Importantly, the Formula also significantly inhibited the growth of PCa xenografts, bone metastasis, and PCSCs activity in vivo by targeting the TAMs/CCL5 pathway. Overall, this study not only elucidates the immunomodulatory mechanism of the FZYL Formula in PCa therapy but also highlights the TAMs/CCL5 axis as a promising therapeutic target.
Collapse
Affiliation(s)
- Chiwei Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Renlun Huang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510000, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Yifeng Zheng
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Jianfu Zhou
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Bowen Yang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Xuan Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Juping Zhang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Bo Pan
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Zhiqiang Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China
| | - Shengqi Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510000, China.
| | - Zhiyu Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510000, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China.
| | - Songtao Xiang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510000, China.
| |
Collapse
|
31
|
El-Derby AM, Khedr MA, Ghoneim NI, Gabr MM, Khater SM, El-Badri N. Plasma-derived extracellular matrix for xenofree and cost-effective organoid modeling for hepatocellular carcinoma. J Transl Med 2024; 22:487. [PMID: 38773585 PMCID: PMC11110239 DOI: 10.1186/s12967-024-05230-7] [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: 01/22/2024] [Accepted: 04/23/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) causes significant cancer mortality worldwide. Cancer organoids can serve as useful disease models by high costs, complexity, and contamination risks from animal-derived products and extracellular matrix (ECM) that limit its applications. On the other hand, synthetic ECM alternatives also have limitations in mimicking native biocomplexity. This study explores the development of a physiologically relevant HCC organoid model using plasma-derived extracellular matrix as a scaffold and nutritive biomatrix with different cellularity components to better mimic the heterogenous HCC microenvironment. Plasma-rich platelet is recognized for its elevated levels of growth factors, which can promote cell proliferation. By employing it as a biomatrix for organoid culture there is a potential to enhance the quality and functionality of organoid models for diverse applications in biomedical research and regenerative medicine and to better replicate the heterogeneous microenvironment of HCC. METHOD To generate the liver cancer organoids, HUH-7 hepatoma cells were cultured alone (homogenous model) or with human bone marrow-derived mesenchymal stromal cells and human umbilical vein endothelial cells (heterogeneous model) in plasma-rich platelet extracellular matrix (ECM). The organoids were grown for 14 days and analyzed for cancer properties including cell viability, invasion, stemness, and drug resistance. RESULTS HCC organoids were developed comprising HUH-7 hepatoma cells with or without human mesenchymal stromal and endothelial cells in plasma ECM scaffolds. Both homogeneous (HUH-7 only) and heterogeneous (mixed cellularity) organoids displayed viability, cancer hallmarks, and chemoresistance. The heterogeneous organoids showed enhanced invasion potential, cancer stem cell populations, and late-stage HCC genetic signatures versus homogeneous counterparts. CONCLUSION The engineered HCC organoids system offers a clinically relevant and cost-effective model to study liver cancer pathogenesis, stromal interactions, and drug resistance. The plasma ECM-based culture technique could enable standardized and reproducible HCC modeling. It could also provide a promising option for organoid culture and scaling up.
Collapse
Affiliation(s)
- Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Mennatallah A Khedr
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Nehal I Ghoneim
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Mahmoud M Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Sherry M Khater
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt.
| |
Collapse
|
32
|
Yu R, Hang Y, Tsai HI, Wang D, Zhu H. Iron metabolism: backfire of cancer cell stemness and therapeutic modalities. Cancer Cell Int 2024; 24:157. [PMID: 38704599 PMCID: PMC11070091 DOI: 10.1186/s12935-024-03329-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024] Open
Abstract
Cancer stem cells (CSCs), with their ability of self-renewal, unlimited proliferation, and multi-directional differentiation, contribute to tumorigenesis, metastasis, recurrence, and resistance to conventional therapy and immunotherapy. Eliminating CSCs has long been thought to prevent tumorigenesis. Although known to negatively impact tumor prognosis, research revealed the unexpected role of iron metabolism as a key regulator of CSCs. This review explores recent advances in iron metabolism in CSCs, conventional cancer therapies targeting iron biochemistry, therapeutic resistance in these cells, and potential treatment options that could overcome them. These findings provide important insights into therapeutic modalities against intractable cancers.
Collapse
Affiliation(s)
- Rong Yu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - Yinhui Hang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China.
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Dongqing Wang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China.
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China.
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
33
|
Zhang K, Hu W, Li F, Wen C, Zhou L, Zhang L, Lian J, Liu S, Wang S, Zhang Y. IL-24 improves efficacy of CAR-T cell therapy by targeting stemness of tumor cells. Br J Cancer 2024; 130:1337-1347. [PMID: 38347092 PMCID: PMC11015030 DOI: 10.1038/s41416-024-02601-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) induce therapeutic resistance and may be an important barrier to cancer immunotherapy. Chimeric antigen receptor T (CAR-T) cell therapy has demonstrated remarkable efficacy in clinical settings. However, CAR-T cell therapy fails in a large proportion of patients, especially in those with solid tumors. It is unclear how CSCs mediate resistance to CAR-T cells, and whether CAR-T cells can more effectively eradicate CSCs. METHODS In this study, the effect of CSCs on CAR-T cell therapy was determined using in vitro and in vivo assays. Subsequently, Interleukin-24 (IL-24) was expressed along with CAR in T cells. Further in vitro and in vivo tests were performed to determine the effects of IL-24 on CSCs and CAR-T cell therapy. RESULTS IL-24 induced apoptosis in CSCs and contributed to T cell activation, differentiation, and proliferation. CAR.IL-24-T cells inhibited CSC enrichment and exhibited stronger antitumor activity in vitro and in vivo. CONCLUSIONS IL-24 helps eliminate CSCs and endows CAR-T cells with improved antitumor reactivity.
Collapse
Affiliation(s)
- Kai Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenhao Hu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Li
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chunli Wen
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lingxiao Zhou
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lei Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingyao Lian
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shasha Liu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shumin Wang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China.
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China.
| |
Collapse
|
34
|
Zhao R, Guo X, Zhang G, Liu S, Ma R, Wang M, Chen S, Zhu W, Liu Y, Gao P, Liu H. CMYC-initiated HNF1A-AS1 overexpression maintains the stemness of gastric cancer cells. Cell Death Dis 2024; 15:288. [PMID: 38654006 PMCID: PMC11039746 DOI: 10.1038/s41419-024-06673-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Cancer stem cells (CSCs) are believed to be responsible for cancer metastasis and recurrence due to their self-renewal ability and resistance to treatment. However, the mechanisms that regulate the stemness of CSCs remain poorly understood. Recently, evidence has emerged suggesting that long non-coding RNAs (lncRNAs) play a crucial role in regulating cancer cell function in different types of malignancies, including gastric cancer (GC). However, the specific means by which lncRNAs regulate the function of gastric cancer stem cells (GCSCs) are yet to be fully understood. In this study, we investigated a lncRNA known as HNF1A-AS1, which is highly expressed in GCSC s and serves as a critical regulator of GCSC stemness and tumorigenesis. Our experiments, both in vitro and in vivo, demonstrated that HNF1A-AS1 maintained the stemness of GC cells. Further analysis revealed that HNF1A-AS1, transcriptionally activated by CMYC, functioned as a competing endogenous RNA by binding to miR-150-5p to upregulate β-catenin expression. This in turn facilitated the entry of β-catenin into the nucleus to activate the Wnt/β-catenin pathway and promote CMYC expression, thereby forming a positive feedback loop that sustained the stemness of GCSCs. We also found that blocking the Wnt/β-catenin pathway effectively inhibited the function of HNF1A-AS1, ultimately resulting in the inhibition of GCSC stemness. Taken together, our results demonstrated that HNF1A-AS1 is a regulator of the stemness of GCSCs and could serve as a potential marker for targeted GC therapy.
Collapse
Affiliation(s)
- Ruinan Zhao
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiangyu Guo
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Guohao Zhang
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Sen Liu
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ranran Ma
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Mengqi Wang
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shiming Chen
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Wenjie Zhu
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yuan Liu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China.
| | - Peng Gao
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Haiting Liu
- Department of Pathology, Qilu Hospital and School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
35
|
Gou Z, Li J, Liu J, Yang N. The hidden messengers: cancer associated fibroblasts-derived exosomal miRNAs as key regulators of cancer malignancy. Front Cell Dev Biol 2024; 12:1378302. [PMID: 38694824 PMCID: PMC11061421 DOI: 10.3389/fcell.2024.1378302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/08/2024] [Indexed: 05/04/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs), a class of stromal cells in the tumor microenvironment (TME), play a key role in controlling cancer cell invasion and metastasis, immune evasion, angiogenesis, and resistance to chemotherapy. CAFs mediate their activities by secreting soluble chemicals, releasing exosomes, and altering the extracellular matrix (ECM). Exosomes contain various biomolecules, such as nucleic acids, lipids, and proteins. microRNA (miRNA), a 22-26 nucleotide non-coding RNA, can regulate the cellular transcription processes. Studies have shown that miRNA-loaded exosomes secreted by CAFs engage in various regulatory communication networks with other TME constituents. This study focused on the roles of CAF-derived exosomal miRNAs in generating cancer malignant characteristics, including immune modulation, tumor growth, migration and invasion, epithelial-mesenchymal transition (EMT), and treatment resistance. This study thoroughly examines miRNA's dual regulatory roles in promoting and suppressing cancer. Thus, changes in the CAF-derived exosomal miRNAs can be used as biomarkers for the diagnosis and prognosis of patients, and their specificity can be used to develop newer therapies. This review also discusses the pressing problems that require immediate attention, aiming to inspire researchers to explore more novel avenues in this field.
Collapse
Affiliation(s)
- Zixuan Gou
- Bethune First Clinical School of Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jianming Liu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Na Yang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
36
|
Sun S, Yang Q, Jiang D, Zhang Y. Nanobiotechnology augmented cancer stem cell guided management of cancer: liquid-biopsy, imaging, and treatment. J Nanobiotechnology 2024; 22:176. [PMID: 38609981 PMCID: PMC11015566 DOI: 10.1186/s12951-024-02432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent both a key driving force and therapeutic target of tumoral carcinogenesis, tumor evolution, progression, and recurrence. CSC-guided tumor diagnosis, treatment, and surveillance are strategically significant in improving cancer patients' overall survival. Due to the heterogeneity and plasticity of CSCs, high sensitivity, specificity, and outstanding targeting are demanded for CSC detection and targeting. Nanobiotechnologies, including biosensors, nano-probes, contrast enhancers, and drug delivery systems, share identical features required. Implementing these techniques may facilitate the overall performance of CSC detection and targeting. In this review, we focus on some of the most recent advances in how nanobiotechnologies leverage the characteristics of CSC to optimize cancer diagnosis and treatment in liquid biopsy, clinical imaging, and CSC-guided nano-treatment. Specifically, how nanobiotechnologies leverage the attributes of CSC to maximize the detection of circulating tumor DNA, circulating tumor cells, and exosomes, to improve positron emission computed tomography and magnetic resonance imaging, and to enhance the therapeutic effects of cytotoxic therapy, photodynamic therapy, immunotherapy therapy, and radioimmunotherapy are reviewed.
Collapse
Affiliation(s)
- Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiang Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
37
|
Wang Y, Wang B, Cao W, Xu X. PTX3 activates POSTN and promotes the progression of glioblastoma via the MAPK/ERK signalling axis. Biochem Biophys Res Commun 2024; 703:149665. [PMID: 38359612 DOI: 10.1016/j.bbrc.2024.149665] [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: 12/24/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Intrinsic brain tumours such as glioblastoma (GBM) are believed to develop from neuroglial stem or progenitor cells. GBM accounts for approximately half of gliomas. GBM has a poor prognosis and a low 5-year survival rate. Pentraxin 3 (PTX3) is overexpressed in GBM, but the potential mechanism is unclear. METHODS Glioblastoma data from the TCGA and CGGA databases were used to analyse PTX3 expression. Subsequently, in vivo and in vitro experiments were conducted to verify the effect of PTX3 silencing in glioma cells on EMT like process and GSC maintenance. The JASPAR database was used to predict the downstream genes of PTX3. POSTN is a novel target gene of PTX3 in gliomas, and this finding was validated using a luciferase reporter gene assay. Western blotting and KEGG enrichment analysis were used to predict the downstream pathway of POSTN, and it was found that the MAPK/ERK pathway might be related to the function of POSTN. RESULTS GBM tissues have higher levels of PTX3 expression than normal brain tissues (NBTs). In functional tests, PTX3 promoted the EMT like process of GBM cells while maintaining the stem cell characteristics of GBM stem cells and enhancing their self-renewal. Moreover, we performed a dual luciferase reporter experiment to confirm that PTX3 binds to the POSTN promoter region. In addition, the expression of key proteins in the MAPK/ERK signalling pathway was increased after PTX3 overexpression. CONCLUSION POSTN is a direct target of PTX3 that promotes GBM growth via the MAPK/ERK signalling pathway.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Binbin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Wenping Cao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
38
|
Zhang Z, Zhang Y. Transcriptional regulation of cancer stem cell: regulatory factors elucidation and cancer treatment strategies. J Exp Clin Cancer Res 2024; 43:99. [PMID: 38561775 PMCID: PMC10986082 DOI: 10.1186/s13046-024-03021-y] [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: 12/04/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer stem cells (CSCs) were first discovered in the 1990s, revealing the mysteries of cancer origin, migration, recurrence and drug-resistance from a new perspective. The expression of pluripotent genes and complex signal regulatory networks are significant features of CSC, also act as core factors to affect the characteristics of CSC. Transcription is a necessary link to regulate the phenotype and potential of CSC, involving chromatin environment, nucleosome occupancy, histone modification, transcription factor (TF) availability and cis-regulatory elements, which suffer from ambient pressure. Especially, the expression and activity of pluripotent TFs are deeply affected by both internal and external factors, which is the foundation of CSC transcriptional regulation in the current research framework. Growing evidence indicates that regulating epigenetic modifications to alter cancer stemness is effective, and some special promoters and enhancers can serve as targets to influence the properties of CSC. Clarifying the factors that regulate CSC transcription will assist us directly target key stem genes and TFs, or hinder CSC transcription through environmental and other related factors, in order to achieve the goal of inhibiting CSC and tumors. This paper comprehensively reviews the traditional aspects of transcriptional regulation, and explores the progress and insights of the impact on CSC transcription and status through tumor microenvironment (TME), hypoxia, metabolism and new meaningful regulatory factors in conjunction with the latest research. Finally, we present opinions on omnidirectional targeting CSCs transcription to eliminate CSCs and address tumor resistance.
Collapse
Affiliation(s)
- Zhengyue Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, People's Republic of China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, People's Republic of China
| | - Yanjie Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, People's Republic of China.
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, People's Republic of China.
| |
Collapse
|
39
|
Wang J, Zhang J, Liu H, Meng L, Gao X, Zhao Y, Wang C, Gao X, Fan A, Cao T, Fan D, Zhao X, Lu Y. N6-methyladenosine reader hnRNPA2B1 recognizes and stabilizes NEAT1 to confer chemoresistance in gastric cancer. Cancer Commun (Lond) 2024; 44:469-490. [PMID: 38512764 PMCID: PMC11024687 DOI: 10.1002/cac2.12534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Chemoresistance is a major cause of treatment failure in gastric cancer (GC). Heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) is an N6-methyladenosine (m6A)-binding protein involved in a variety of cancers. However, whether m6A modification and hnRNPA2B1 play a role in GC chemoresistance is largely unknown. In this study, we aimed to investigate the role of hnRNPA2B1 and the downstream mechanism in GC chemoresistance. METHODS The expression of hnRNPA2B1 among public datasets were analyzed and validated by quantitative PCR (qPCR), Western blotting, immunofluorescence, and immunohistochemical staining. The biological functions of hnRNPA2B1 in GC chemoresistance were investigated both in vitro and in vivo. RNA sequencing, methylated RNA immunoprecipitation, RNA immunoprecipitation, and RNA stability assay were performed to assess the association between hnRNPA2B1 and the binding RNA. The role of hnRNPA2B1 in maintenance of GC stemness was evaluated by bioinformatic analysis, qPCR, Western blotting, immunofluorescence, and sphere formation assays. The expression patterns of hnRNPA2B1 and downstream regulators in GC specimens from patients who received adjuvant chemotherapy were analyzed by RNAscope and multiplex immunohistochemistry. RESULTS Elevated expression of hnRNPA2B1 was found in GC cells and tissues, especially in multidrug-resistant (MDR) GC cell lines. The expression of hnRNPA2B1 was associated with poor outcomes of GC patients, especially in those who received 5-fluorouracil treatment. Silencing hnRNPA2B1 effectively sensitized GC cells to chemotherapy by inhibiting cell proliferation and inducing apoptosis both in vitro and in vivo. Mechanically, hnRNPA2B1 interacted with and stabilized long noncoding RNA NEAT1 in an m6A-dependent manner. Furthermore, hnRNPA2B1 and NEAT1 worked together to enhance the stemness properties of GC cells via Wnt/β-catenin signaling pathway. In clinical specimens from GC patients subjected to chemotherapy, the expression levels of hnRNPA2B1, NEAT1, CD133, and CD44 were markedly elevated in non-responders compared with responders. CONCLUSION Our findings indicated that hnRNPA2B1 interacts with and stabilizes lncRNA NEAT1, which contribute to the maintenance of stemness property via Wnt/β-catenin pathway and exacerbate chemoresistance in GC.
Collapse
Affiliation(s)
- Jiayao Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
- The Air Force Hospital of Southern Theater CommandGuangzhouGuangdongP. R. China
| | - Jiehao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
- The Air Force Hospital of Southern Theater CommandGuangzhouGuangdongP. R. China
| | - Hao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Lingnan Meng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
- National Center for International Research of Bio‐targeting TheranosticsGuangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNanningGuangxiP. R. China
| | - Xianchun Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Yihan Zhao
- Second Clinical CollegeShaanxi University of Traditional Chinese MedicineXianyangShaanxiP. R. China
| | - Chen Wang
- College of Life SciencesNorthwest UniversityXi'anShaanxiP. R. China
| | - Xiaoliang Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Ahui Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Tianyu Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anShaanxiP. R. China
| |
Collapse
|
40
|
Sato T, Oshi M, Huang JL, Chida K, Roy AM, Endo I, Takabe K. CD133 expression is associated with less DNA repair, better response to chemotherapy and survival in ER-positive/HER2-negative breast cancer. RESEARCH SQUARE 2024:rs.3.rs-4148608. [PMID: 38585981 PMCID: PMC10996805 DOI: 10.21203/rs.3.rs-4148608/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Purpose CD133, a cancer stem cells (CSC) marker, has been reported to be associated with treatment resistance and worse survival in triple-negative breast cancer (BC). However, the clinical relevance of CD133 expression in ER-positive/HER2-negative (ER+/HER2-) BC, the most abundant subtype, remains unknown. Methods The BC cohorts from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC, n = 1904) and The Cancer Genome Atlas (TCGA, n = 1065) were used to obtain biological variables and gene expression data. Results Epithelial cells were the exclusive source of CD133 gene expression in a bulk BC. CD133-high ER+/HER2- BC was associated with CD24, NOTCH1, DLL1, and ALDH1A1 gene expressions, as well as with WNT/β-Catenin, Hedgehog, and Notchsignaling pathways, all characteristic for CSC. Consistent with a CSC phenotype, CD133-low BC was enriched with gene sets related to cell proliferation, such as G2M Checkpoint, MYC Targets V1, E2F Targets, and Ki67 gene expression. CD133-low BC was also linked with enrichment of genes related to DNA repair, such as BRCA1, E2F1, E2F4, CDK1/2. On the other hand, CD133-high tumors had proinflammatory microenvironment, higher activity of immune cells, and higher expression of genes related to inflammation and immune response. Finally, CD133-high tumors had better pathological complete response after neoadjuvant chemotherapy in GSE25066 cohort and better disease-free survival and overall survival in both TCGA and METABRIC cohorts. Conclusion CD133-high ER+/HER2- BC was associated with CSC phenotype such as less cell proliferation and DNA repair, but also with enhanced inflammation, better response to neoadjuvant chemotherapy and better prognosis.
Collapse
Affiliation(s)
| | - Masanori Oshi
- Yokohama City University Graduate School of Medicine
| | | | | | | | - Itaru Endo
- Yokohama City University Graduate School of Medicine
| | | |
Collapse
|
41
|
Picher EA, Wahajuddin M, Barth S, Chisholm J, Shipley J, Pors K. The Capacity of Drug-Metabolising Enzymes in Modulating the Therapeutic Efficacy of Drugs to Treat Rhabdomyosarcoma. Cancers (Basel) 2024; 16:1012. [PMID: 38473371 DOI: 10.3390/cancers16051012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Rhabdomyosarcoma (RMS) is a rare soft tissue sarcoma (STS) that predominantly affects children and teenagers. It is the most common STS in children (40%) and accounts for 5-8% of total childhood malignancies. Apart from surgery and radiotherapy in eligible patients, standard chemotherapy is the only therapeutic option clinically available for RMS patients. While survival rates for this childhood cancer have considerably improved over the last few decades for low-risk and intermediate-risk cases, the mortality rate remains exceptionally high in high-risk RMS patients with recurrent and/or metastatic disease. The intensification of chemotherapeutic protocols in advanced-stage RMS has historically induced aggravated toxicity with only very modest therapeutic gain. In this review, we critically analyse what has been achieved so far in RMS therapy and provide insight into how a diverse group of drug-metabolising enzymes (DMEs) possess the capacity to modify the clinical efficacy of chemotherapy. We provide suggestions for new therapeutic strategies that exploit the presence of DMEs for prodrug activation, targeted chemotherapy that does not rely on DMEs, and RMS-molecular-subtype-targeted therapies that have the potential to enter clinical evaluation.
Collapse
Affiliation(s)
- Enric Arasanz Picher
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Julia Chisholm
- Children and Young People's Unit, Royal Marsden Hospital, Institute of Cancer Research, Sutton SM2 5PR, UK
| | - Janet Shipley
- Sarcoma Molecular Pathology Group, Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Klaus Pors
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
42
|
Yuan Y, Zhang XF, Li YC, Chen HQ, Wen T, Zheng JL, Zhao ZY, Hu QY. VX-509 attenuates the stemness characteristics of colorectal cancer stem-like cells by regulating the epithelial-mesenchymal transition through Nodal/Smad2/3 signaling. World J Stem Cells 2024; 16:207-227. [PMID: 38455101 PMCID: PMC10915959 DOI: 10.4252/wjsc.v16.i2.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/19/2023] [Accepted: 01/16/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Colorectal cancer stem cells (CCSCs) are heterogeneous cells that can self-renew and undergo multidirectional differentiation in colorectal cancer (CRC) patients. CCSCs are generally accepted to be important sources of CRC and are responsible for the progression, metastasis, and therapeutic resistance of CRC. Therefore, targeting this specific subpopulation has been recognized as a promising strategy for overcoming CRC. AIM To investigate the effect of VX-509 on CCSCs and elucidate the underlying mechanism. METHODS CCSCs were enriched from CRC cell lines by in conditioned serum-free medium. Western blot, Aldefluor, transwell and tumorigenesis assays were performed to verify the phenotypic characteristics of the CCSCs. The anticancer efficacy of VX-509 was assessed in HCT116 CCSCs and HT29 CCSCs by performing cell viability analysis, colony formation, sphere formation, flow cytometry, and western blotting assessments in vitro and tumor growth, immunohistochemistry and immunofluorescence assessments in vivo. RESULTS Compared with parental cells, sphere cells derived from HCT116 and HT29 cells presented increased expression of stem cell transcription factors and stem cell markers and were more potent at promoting migration and tumorigenesis, demonstrating that the CRC sphere cells displayed CSC features. VX-509 inhibited the tumor malignant biological behavior of CRC-stem-like cells, as indicated by their proliferation, migration and clonality in vitro, and suppressed the tumor of CCSC-derived xenograft tumors in vivo. Besides, VX-509 suppressed the CSC characteristics of CRC-stem-like cells and inhibited the progression of epithelial-mesenchymal transition (EMT) signaling in vitro. Nodal was identified as the regulatory factor of VX-509 on CRC stem-like cells through analyses of differentially expressed genes and CSC-related database information. VX-509 markedly downregulated the expression of Nodal and its downstream phosphorylated Smad2/3 to inhibit EMT progression. Moreover, VX-509 reversed the dedifferentiation of CCSCs and inhibited the progression of EMT induced by Nodal overexpression. CONCLUSION VX-509 prevents the EMT process in CCSCs by inhibiting the transcription and protein expression of Nodal, and inhibits the dedifferentiated self-renewal of CCSCs.
Collapse
Affiliation(s)
- Yun Yuan
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Xu-Fan Zhang
- Department of Nuclear Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Yu-Chen Li
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Hong-Qing Chen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Tian Wen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Jia-Lian Zheng
- Department of Hepatology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning Province, China
| | - Zi-Yi Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu 610072, Sichuan Province, China
| | - Qiong-Ying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China.
| |
Collapse
|
43
|
Huang JZ, Li JD, Chen G, He RQ. Identification of the key genes and mechanisms associated with transcatheter arterial chemoembolisation refractoriness in hepatocellular carcinoma. World J Clin Oncol 2024; 15:62-88. [PMID: 38292662 PMCID: PMC10823944 DOI: 10.5306/wjco.v15.i1.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Transcatheter arterial embolisation (TACE) is the primary treatment for intermediate-stage hepatocellular carcinoma (HCC) patients while some HCC cases have shown resistance to TACE. AIM To investigate the key genes and potential mechanisms correlated with TACE refractoriness in HCC. METHODS The microarray datasets of TACE-treated HCC tissues, HCC and non-HCC tissues were collected by searching multiple public databases. The respective differentially expressed genes (DEGs) were attained via limma R package. Weighted gene co-expression network analysis was employed for identifying the significant modules related to TACE non-response. TACE refractoriness-related genes were obtained by intersecting up-regulated TACE-associated and HCC-associated DEGs together with the genes in significant modules related to TACE non-response. The key genes expression in the above two pairs of samples was compared respectively via Wilcoxon tests and standard mean differences model. The prognostic value of the key genes was evaluated by Kaplan-Meier curve. Multivariate analysis was utilised to investigate the independent prognostic factor in key genes. Single-cell RNA (scRNA) sequencing analysis was conducted to explore the cell types in HCC. TACE refractoriness-related genes activity was calculated via AUCell packages. The CellChat R package was used for the investigation of the cell-cell communication between the identified cell types. RESULTS HCC tissues of TACE non-responders (n = 66) and TACE responders (n = 81), HCC (n = 3941) and non-HCC (n = 3443) tissues were obtained. The five key genes, DLG associated protein 5 (DLGAP5), Kinesin family member 20A (KIF20A), Assembly factor for spindle microtubules (ASPM), Kinesin family member 11 (KIF11) and TPX2 microtubule nucleation factor (TPX2) in TACE refractoriness-related genes, were identified. The five key genes were all up-regulated in the TACE non-responders group and the HCC group. High expression of the five key genes predicted poor prognosis in HCC. Among the key genes, TPX2 was an independent prognostic factor. Four cell types, hepatocytes, embryonic stem cells, T cells and B cells, were identified in the HCC tissues. The TACE refractoriness-related genes expressed primarily in hepatocytes and embryonic stem cells. Hepatocytes, as the providers of ligands, had the strongest interaction with embryonic stem cells that provided receptors. CONCLUSION Five key genes (DLGAP5, KIF20A, ASPM, KIF11 and TPX2) were identified as promoting refractory TACE. Hepatocytes and embryonic stem cells were likely to boost TACE refractoriness.
Collapse
Affiliation(s)
- Jie-Zhuang Huang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Di Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
44
|
Wang N, Ma JM. Progress of Cancer Stem Cells in Retinoblastoma. Curr Stem Cell Res Ther 2024; 19:1093-1101. [PMID: 37815190 DOI: 10.2174/011574888x252989230921065809] [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: 04/09/2023] [Revised: 07/10/2023] [Accepted: 07/24/2023] [Indexed: 10/11/2023]
Abstract
The theory of cancer stem cells is a breakthrough discovery that offers exciting possibilities for comprehending the biological behavior of tumors. More and more evidence suggests that retinoblastoma cancer stem cells promote tumor growth and are likely to be the origin of tumor formation, drug resistance, recurrence, and metastasis. At present, some progress has been made in the verification, biological behavior, and drug resistance mechanism of retinoblastoma cancer stem cells. This article aims to review the relevant research and explore future development direction.
Collapse
Affiliation(s)
- Nan Wang
- Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jian-Min Ma
- Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
45
|
Wang WD, Guo YY, Yang ZL, Su GL, Sun ZJ. Sniping Cancer Stem Cells with Nanomaterials. ACS NANO 2023; 17:23262-23298. [PMID: 38010076 DOI: 10.1021/acsnano.3c07828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cancer stem cells (CSCs) drive tumor initiation, progression, and therapeutic resistance due to their self-renewal and differentiation capabilities. Despite encouraging progress in cancer treatment, conventional approaches often fail to eliminate CSCs, necessitating the development of precise targeted strategies. Recent advances in materials science and nanotechnology have enabled promising CSC-targeted approaches, harnessing the power of tailoring nanomaterials in diverse therapeutic applications. This review provides an update on the current landscape of nanobased precision targeting approaches against CSCs. We elucidate the nuanced application of organic, inorganic, and bioinspired nanomaterials across a spectrum of therapeutic paradigms, encompassing targeted therapy, immunotherapy, and multimodal synergistic therapies. By examining the accomplishments and challenges in this potential field, we aim to inform future efforts to advance nanomaterial-based therapies toward more effective "sniping" of CSCs and tumor clearance.
Collapse
Affiliation(s)
- Wen-Da Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Yan-Yu Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhong-Lu Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Guang-Liang Su
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
46
|
Tran DN, Hoang TTH, Nandanwar S, Ho VTTX, Pham VT, Vu HD, Nguyen XH, Nguyen HT, Nguyen TV, Nguyen TKV, Tran DL, Park M, Lee S, Pham TC. Dual anticancer and antibacterial activity of fluorescent naphthoimidazolium salts. RSC Adv 2023; 13:36430-36438. [PMID: 38099251 PMCID: PMC10719908 DOI: 10.1039/d3ra06555c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023] Open
Abstract
Cancer has emerged as a significant global health challenge, ranking as the second leading cause of death worldwide. Moreover, cancer patients frequently experience compromised immune systems, rendering them susceptible to bacterial infections. Combining anticancer and antibacterial properties in a single drug could lead to improved overall treatment outcomes and patient well-being. In this context, the present study focused on a series of hydrophilic naphthoimidazolium salts with donor groups (NI-R), aiming to create dual-functional agents with antibacterial and anticancer activities. Among these compounds, NI-TPA demonstrated notable antibacterial activity, particularly against drug-resistant bacteria, with MIC value of 7.8 μg mL-1. Furthermore, NI-TPA exhibited the most potent cytotoxicity against four different cancer cell lines, with an IC50 range of 0.67-2.01 μg mL-1. The observed high cytotoxicity of NI-TPA agreed with molecular docking and dynamic simulation studies targeting c-Met kinase protein. Additionally, NI-TPA stood out as the most promising candidate for two-photo excitation, fluorescence bioimaging, and localization in lysosomes. The study findings open new avenues for the design and development of imidazolium salts that could be employed in phototheranostic applications for cancer treatment and bacterial infections.
Collapse
Affiliation(s)
- Dung Ngoc Tran
- Faculty of Chemistry, Hanoi National University of Education Hanoi Vietnam
| | | | - Sondavid Nandanwar
- Eco-friendly New Materials Research Center, Korea Research Institute of Chemical Technology 141 Gajeong-ro, Yuseong-gu Daejeon City Republic of Korea
| | | | - Van Thong Pham
- R&D Center, Vietnam Education and Technology Transfer JSC Cau Giay Hanoi Vietnam
| | - Huy Duc Vu
- Department of Radiology, School of Medicine, Daegu Catholic University Daegu 42472 Korea
| | - Xuan Ha Nguyen
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology 18 Hoang Quoc Viet, Cau Giay Hanoi Vietnam
| | - Huy Trung Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology 18 Hoang Quoc Viet, Cau Giay Hanoi Vietnam
| | - Trang Van Nguyen
- Institute for Tropical Technology, Vietnam Academy of Science and Technology 18 Hoang Quoc Viet, Cau Giay Hanoi Vietnam
| | - Thuy Kieu Van Nguyen
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University Busan 48513 Korea
| | - Dai Lam Tran
- Institute for Tropical Technology, Vietnam Academy of Science and Technology 18 Hoang Quoc Viet, Cau Giay Hanoi Vietnam
| | - Myeongkee Park
- Department of Chemistry, Pukyong National University Busan 48513 Korea
| | - Songyi Lee
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University Busan 48513 Korea
- Department of Chemistry, Pukyong National University Busan 48513 Korea
| | - Thanh Chung Pham
- Institute for Tropical Technology, Vietnam Academy of Science and Technology 18 Hoang Quoc Viet, Cau Giay Hanoi Vietnam
| |
Collapse
|
47
|
Yang X, Deng L, Diao X, Yang S, Zou L, Yang Q, Li J, Nie J, Zhao L, Jiao B. Targeting cuproptosis by zinc pyrithione in triple-negative breast cancer. iScience 2023; 26:108218. [PMID: 37953954 PMCID: PMC10637938 DOI: 10.1016/j.isci.2023.108218] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/07/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Triple-negative breast cancer (TNBC) poses a considerable challenge due to its aggressive nature. Notably, metal ion-induced cell death, such as ferroptosis, has garnered significant attention and demonstrated potential implications for cancer. Recently, cuproptosis, a potent cell death pathway reliant on copper, has been identified. However, whether cuproptosis can be targeted for cancer treatment remains uncertain. Here, we screened the US Food and Drug Administration (FDA)-approved drug library and identified zinc pyrithione (ZnPT) as a compound that significantly inhibited TNBC progression. RNA sequencing revealed that ZnPT disrupted copper homeostasis. Furthermore, ZnPT facilitated the oligomerization of dihydrolipoamide S-acetyltransferase, a landmark molecule of cuproptosis. Clinically, high expression levels of cuproptosis-related proteins were significantly correlated with poor prognosis in TNBC patients. Collectively, these findings indicate that ZnPT can induce cell death by targeting and disrupting copper homeostasis, providing a potential experimental foundation for exploring cuproptosis as a target in drug discovery for TNBC patients.
Collapse
Affiliation(s)
- Xu Yang
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650201, China
| | - Li Deng
- Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China
| | - Xianhong Diao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650201, China
| | - Siyuan Yang
- Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China
| | - Li Zou
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Qin Yang
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Jian Li
- Institutional Center for Shared Technologies and Facilities, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Jianyun Nie
- Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China
| | - Lina Zhao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Baowei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650203, China
| |
Collapse
|
48
|
Doustmihan A, Fathi M, Mazloomi M, Salemi A, Hamblin MR, Jahanban-Esfahlan R. Molecular targets, therapeutic agents and multitasking nanoparticles to deal with cancer stem cells: A narrative review. J Control Release 2023; 363:57-83. [PMID: 37739017 DOI: 10.1016/j.jconrel.2023.09.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
There is increasing evidence that malignant tumors are initiated and maintained by a sub-population of tumor cells that have similar biological properties to normal adult stem cells. This very small population of Cancer Stem Cells (CSC) comprises tumor initiating cells responsible for cancer recurrence, drug resistance and metastasis. Conventional treatments such as chemotherapy, radiotherapy and surgery, in addition to being potentially toxic and non-specific, may paradoxically increase the population, spread and survival of CSCs. Next-generation sequencing and omics technologies are increasing our understanding of the pathways and factors involved in the development of CSCs, and can help to discover new therapeutic targets against CSCs. In addition, recent advances in nanomedicine have provided hope for the development of optimal specific therapies to eradicate CSCs. Moreover, the use of artificial intelligence and nano-informatics can elucidate new drug targets, and help to design drugs and nanoparticles (NPs) to deal with CSCs. In this review, we first summarize the properties of CSCs and describe the signaling pathways and molecular characteristics responsible for the emergence and survival of CSCs. Also, the location of CSCs within the tumor and the effect of host factors on the creation and maintenance of CSCs are discussed. Newly discovered molecular targets involved in cancer stemness and some novel therapeutic compounds to combat CSCs are highlighted. The optimum properties of anti-CSC NPs, including blood circulation and stability, tumor accumulation and penetration, cellular internalization, drug release, endosomal escape, and aptamers designed for specific targeting of CSCs are covered. Finally, some recent smart NPs designed for therapeutic and theranostic purposes to overcome CSCs are discussed.
Collapse
Affiliation(s)
- Abolfazl Doustmihan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - MirAhmad Mazloomi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysan Salemi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Zhong Q, Wang H, Yang J, Tu R, Li A, Zeng G, Zheng Q, Yu Liu Z, Shang‐Guan Z, Bo Huang X, Huang Q, Li Y, Zheng H, Lin G, Huang Z, Xu K, Qiu W, Jiang M, Zhao Y, Lin J, Huang Z, Huang J, Li P, Xie J, Zheng C, Chen Q, Huang C. Loss of ATOH1 in Pit Cell Drives Stemness and Progression of Gastric Adenocarcinoma by Activating AKT/mTOR Signaling through GAS1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301977. [PMID: 37824217 PMCID: PMC10646280 DOI: 10.1002/advs.202301977] [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: 03/29/2023] [Revised: 08/19/2023] [Indexed: 10/14/2023]
Abstract
Gastric cancer stem cells (GCSCs) are self-renewing tumor cells that govern chemoresistance in gastric adenocarcinoma (GAC), whereas their regulatory mechanisms remain elusive. Here, the study aims to elucidate the role of ATOH1 in the maintenance of GCSCs. The preclinical model and GAC sample analysis indicate that ATOH1 deficiency is correlated with poor GAC prognosis and chemoresistance. ScRNA-seq reveals that ATOH1 is downregulated in the pit cells of GAC compared with those in paracarcinoma samples. Lineage tracing reveals that Atoh1 deletion strongly confers pit cell stemness. ATOH1 depletion significantly accelerates cancer stemness and chemoresistance in Tff1-CreERT2; Rosa26Tdtomato and Tff1-CreERT2; Apcfl/fl ; p53fl/fl (TcPP) mouse models and organoids. ATOH1 deficiency downregulates growth arrest-specific protein 1 (GAS1) by suppressing GAS1 promoter transcription. GAS1 forms a complex with RET, which inhibits Tyr1062 phosphorylation, and consequently activates the RET/AKT/mTOR signaling pathway by ATOH1 deficiency. Combining chemotherapy with drugs targeting AKT/mTOR signaling can overcome ATOH1 deficiency-induced chemoresistance. Moreover, it is confirmed that abnormal DNA hypermethylation induces ATOH1 deficiency. Taken together, the results demonstrate that ATOH1 loss promotes cancer stemness through the ATOH1/GAS1/RET/AKT/mTOR signaling pathway in GAC, thus providing a potential therapeutic strategy for AKT/mTOR inhibitors in GAC patients with ATOH1 deficiency.
Collapse
|
50
|
Fan H, Song L, Fan J, Ma J, Li X, Zhang J, Hu J, Wu Z, Zhang D, Wang L. Decoding meningioma heterogeneity and neoplastic cell-macrophage interaction through single-cell transcriptome profiling across pathological grades. J Transl Med 2023; 21:751. [PMID: 37880655 PMCID: PMC10599053 DOI: 10.1186/s12967-023-04445-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/16/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Analyzing meningioma of distinct pathological types at the single-cell level can provide new and valuable insights into the specific biological mechanisms of each cellular subpopulation, as well as their vital interplay within the tumor microenvironment. METHODS We recruited patients diagnosed with four distinct types of meningioma and performed single-cell RNA sequencing on their tumor samples, concurrently analyzing a publicly available dataset for comparison. Next, we separated the cells into discrete clusters and identified their unique identities. Using pseudotime analysis, we demonstrated cellular differentiation and dynamics. To investigate biological function, we employed weighted gene co-expression network analysis, gene regulatory network, and gene set enrichment analysis. Additionally, we conducted cell-cell communication analyses to characterize interactions among different clusters and validated a crucial interaction using multiple immunofluorescence staining. RESULTS The single-cell transcriptomic profiles for five meningioma of different pathological types demonstrated that neoplastic cells exhibited high inter-sample heterogeneity and diverse biological functions featured by metabolic regulation. A small cluster of neoplastic cells (N5 cluster, < 3%) was most proliferative, indicated by high expression of MKI67 and TOP2A. They were primarily observed in our atypical and transitional meningioma samples and located at the beginning of the pseudotime differentiation branch for neoplastic cells. Macrophages, the most abundant immune cells present, showed two distinct developmental trajectories, one promoting and the other suppressing meningioma growth, with the MIF-CD74 interaction serving as the primary signaling pathway for MIF signals in the tumor environment. Unexpectedly, despite its small cluster size, the N5 cluster demonstrated a significant contribution in this interaction. By staining pathological sections of more samples, we found that this interaction was widely present in different types of meningiomas. CONCLUSIONS Meningioma neoplastic cells' diverse types cause inter-sample heterogeneity and a wide range of functions. Some proliferative neoplastic cell may educate macrophages, which promotes tumorigenesis possibly through the MIF-CD74 interaction. It provides novel clues for future potential therapeutic avenues.
Collapse
Affiliation(s)
- Hailang Fan
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Lairong Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Jian Fan
- Department of Urology, Peking University First Hospital, Institute of Urology, National Urological Cancer Center, Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, 100871, China
| | - Junpeng Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiaojie Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054-1901, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77225-0334, USA
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Dake Zhang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, 100191, China.
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|