1
|
Li Y, Guo T, He J, Liu D, Peng S, Xu A. SLC35A2-mediated bisected GlcNAc-modified extracellular vesicles enhance immune regulation in breast cancer lung metastasis. Int Immunopharmacol 2025; 154:114505. [PMID: 40157085 DOI: 10.1016/j.intimp.2025.114505] [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: 12/07/2024] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
This study investigates the role of SLC35A2-mediated bisected GlcNAc-modified small extracellular vesicles (sEVs) in breast cancer (BC) lung metastasis. By modulating B3GALT1 expression, these sEVs regulate the pre-metastatic immune microenvironment, enhancing CD8+ T cell infiltration and reducing immune evasion. The use of β-peptide-loaded sEVs further amplifies anti-metastatic effects, as demonstrated in vivo mouse models and molecular analyses. These findings underscore the therapeutic potential of glycosylation-modified sEVs in enhancing immune responses and controlling BC metastasis.
Collapse
Affiliation(s)
- Yangyang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Tao Guo
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Juntong He
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Defeng Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Shihao Peng
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Aman Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
2
|
Wang J, Bao S, An Q, Li C, Feng J. Roles of extracellular vesicles from different origins in metabolic-associated fatty liver disease: progress and perspectives. Front Immunol 2025; 16:1544012. [PMID: 40129979 PMCID: PMC11930831 DOI: 10.3389/fimmu.2025.1544012] [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: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/26/2025] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is the most common chronic liver disease worldwide, associated with systemic metabolic dysregulation. It can progress from simple hepatic steatosis (MAFL) to more severe conditions like Metabolic-Associated Steatohepatitis (MASH), fibrosis, cirrhosis, and Hepatocellular Carcinoma (HCC). There is a critical lack of reliable non-invasive diagnostic methods and effective pharmaceutical treatments for MAFLD/MASH, emphasizing the need for further research. Extracellular vesicles (EVs) are nanoscale structures that play important roles in cell signaling by delivering bioactive molecules. However, there is a significant gap in literature regarding the roles of EVs from hosts, plants, and microbiota in MAFLD. This review explores the potential of EVs from various sources-host, plants, and microbiota-as biomarkers, therapeutic agents, drug carriers, and treatment targets for MAFLD. Firstly, the roles of host-derived extracellular vesicles (EVs) in MAFLD, with a focus on cell-type specific EVs and their components-proteins, miRNAs, and lipids-for disease diagnosis and monitoring were discussed. Moreover, it highlighted the therapeutic potential of mesenchymal stem cell (MSC)-derived EVs in reducing lipid accumulation and liver injury, and immune cell-derived EVs in mitigating inflammation and fibrosis. The review also discussed the use of host-derived EVs as drug carriers and therapeutic targets due to their ability to deliver bioactive molecules that impact disease mechanisms. Additionally, it summarized research on plant-derived EVs, which help reduce liver lipid accumulation, inflammation, and enhance gut barrier function in MAFLD. Also, the review explored microbial-derived EVs as novel therapeutic targets, particularly in relation to insulin resistance, liver inflammation, and dysfunction in MAFLD. Overall, by exploring the diverse roles of EVs from host, plant, and microbiota sources in MAFLD, this review offers valuable insights into their potential as non-invasive biomarkers and novel therapeutic strategies, which could pave the way for more effective diagnostic and treatment options for this increasingly prevalent liver disease. Notably, the challenges of translating EVs into clinical practice were also thoroughly discussed, aiming to provide possible directions and strategies for future research.
Collapse
Affiliation(s)
- Jing Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shuoqiang Bao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qi An
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Caihong Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| |
Collapse
|
3
|
Mehrvar A, Akbari M, Khosroshahi EM, Nekavand M, Mokhtari K, Baniasadi M, Aghababaian M, Karimi M, Amiri S, Moazen A, Maghsoudloo M, Alimohammadi M, Rahimzadeh P, Farahani N, Vaghar ME, Entezari M, Hashemi M. The impact of exosomes on bone health: A focus on osteoporosis. Pathol Res Pract 2024; 263:155618. [PMID: 39362132 DOI: 10.1016/j.prp.2024.155618] [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: 08/29/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Osteoporosis is a widespread chronic condition. Although standard treatments are generally effective, they are frequently constrained by side effects and the risk of developing drug resistance. A promising area of research is the investigation of extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, which play a crucial role in bone metabolism. Exosomes, in particular, have shown significant potential in both the diagnosis and treatment of osteoporosis. EVs derived from osteoclasts, osteoblasts, mesenchymal stem cells, and other sources can influence bone metabolism, while exosomes from inflammatory and tumor cells may exacerbate bone loss, highlighting their dual role in osteoporosis pathology. This review offers a comprehensive overview of EV biogenesis, composition, and function in osteoporosis, focusing on their diagnostic and therapeutic potential. We examine the roles of various types of EVs and their cargo-proteins, RNAs, and lipids-in bone metabolism. Additionally, we explore the emerging applications of EVs as biomarkers and therapeutic agents, emphasizing the need for further research to address current challenges and enhance EV-based strategies for managing osteoporosis.
Collapse
Affiliation(s)
- Amir Mehrvar
- Assistant Professor, Department of Orthopedics, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadarian Akbari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrandokht Nekavand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Midwifery, Faculty of nursing and midwifery, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mojtaba Baniasadi
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran; MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Aghababaian
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Karimi
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- MD, Assistant Professor of Orthopaedic Surgery, Shohadaye Haftom-e-Tir Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Moazen
- Department of Orthopedics, Bone and Joint Reconstruction Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad Eslami Vaghar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of gynecology, Faculty of Medicine, Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Zhang J, Lai Z, Zhang Z, Zheng S. Fluorescent monitoring osteogenic differentiation of osteosarcoma cells with an aggregation-induced emission probe. Heliyon 2024; 10:e31664. [PMID: 38828353 PMCID: PMC11140698 DOI: 10.1016/j.heliyon.2024.e31664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
Osteosarcoma is widely believed to be an osteogenic differentiation disorder. In recent years, to further understand this disease, a lot resources were poured into the potential link between differentiation defects and tumorigenesis. Long-term monitoring of the differentiation progress of osteosarcoma cells is of great importance. In order to better promote the research, we have developed a novel fluorescent probe called PTB-EDTA, which exhibits remarkable bio-compatibility and demonstrates high selectivity towards osteosarcoma cells. Not only is the PTB-EDTA is capable of live cell imaging while conventional histology requires to kill the cells, its fluorescence is also enhanced as the osteogenic differentiation proceeding. These properties make PTB-EDTA a promising tool for monitoring osteosarcoma cells.
Collapse
Affiliation(s)
- Junxiong Zhang
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| | - Zhongming Lai
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| | - Zhongmin Zhang
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| | - Shuai Zheng
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| |
Collapse
|
5
|
Griffin K, Mizenko R, Arun V, Carney RP, Leach JK. Extracellular Vesicles from Highly Metastatic Osteosarcoma Cells Induce Pro-Tumorigenic Macrophage Phenotypes. Adv Biol (Weinh) 2024; 8:e2300577. [PMID: 38596830 PMCID: PMC11178448 DOI: 10.1002/adbi.202300577] [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: 10/25/2023] [Revised: 03/27/2024] [Indexed: 04/11/2024]
Abstract
Metastasis is the principal factor in poor prognosis for individuals with osteosarcoma (OS). Understanding the events that lead to metastasis is critical to develop better interventions for this disease. Alveolar macrophages are potentially involved in priming the lung microenvironment for OS metastasis, yet the mechanisms involved in this process remain unclear. Since extracellular vesicles (EVs) are a known actor in primary tumor development, their potential role in OS metastagenesis through macrophage modulation is explored here. The interaction of EVs isolated from highly metastatic (K7M2) and less metastatic (K12) osteosarcoma cell lines is compared with a peritoneal macrophage cell line. An EV concentration that reproducibly induced macrophage migration is identified first, then used for later experiments. By confocal microscopy, both EV types associated with M0 or M1 macrophages; however, only K7M2-EVs are associated with M2 macrophages, an interaction that is abrogated by EV pre-treatment with anti-CD47 antibody. Interestingly, all interactions appeared to be surface binding, not internalized. In functional studies, K7M2-EVs polarized fewer macrophages to M1. Together, these data suggest that K7M2-EVs have unique interactions with macrophages that can contribute to the production of a higher proportion of pro-tumor type macrophages, thereby accelerating metastasis.
Collapse
Affiliation(s)
- Katherine Griffin
- School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| | - Rachel Mizenko
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - Vishalakshi Arun
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - Randy P. Carney
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - J. Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, California, USA
| |
Collapse
|
6
|
Singhto N, Pongphitcha P, Jinawath N, Hongeng S, Chutipongtanate S. Extracellular Vesicles for Childhood Cancer Liquid Biopsy. Cancers (Basel) 2024; 16:1681. [PMID: 38730633 PMCID: PMC11083250 DOI: 10.3390/cancers16091681] [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: 04/09/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Liquid biopsy involves the utilization of minimally invasive or noninvasive techniques to detect biomarkers in biofluids for disease diagnosis, monitoring, or guiding treatments. This approach is promising for the early diagnosis of childhood cancer, especially for brain tumors, where tissue biopsies are more challenging and cause late detection. Extracellular vesicles offer several characteristics that make them ideal resources for childhood cancer liquid biopsy. Extracellular vesicles are nanosized particles, primarily secreted by all cell types into body fluids such as blood and urine, and contain molecular cargos, i.e., lipids, proteins, and nucleic acids of original cells. Notably, the lipid bilayer-enclosed structure of extracellular vesicles protects their cargos from enzymatic degradation in the extracellular milieu. Proteins and nucleic acids of extracellular vesicles represent genetic alterations and molecular profiles of childhood cancer, thus serving as promising resources for precision medicine in cancer diagnosis, treatment monitoring, and prognosis prediction. This review evaluates the recent progress of extracellular vesicles as a liquid biopsy platform for various types of childhood cancer, discusses the mechanistic roles of molecular cargos in carcinogenesis and metastasis, and provides perspectives on extracellular vesicle-guided therapeutic intervention. Extracellular vesicle-based liquid biopsy for childhood cancer may ultimately contribute to improving patient outcomes.
Collapse
Affiliation(s)
- Nilubon Singhto
- Ramathibodi Comprehensive Cancer Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Pongpak Pongphitcha
- Bangkok Child Health Center, Bangkok Hospital Headquarters, Bangkok 10130, Thailand;
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
- Integrative Computational Biosciences Center, Mahidol University, Nakon Pathom 73170, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Somchai Chutipongtanate
- MILCH and Novel Therapeutics Laboratory, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Extracellular Vesicle Working Group, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
7
|
Yu T, Jiang W, Wang Y, Zhou Y, Jiao J, Wu M. Chimeric antigen receptor T cells in the treatment of osteosarcoma (Review). Int J Oncol 2024; 64:40. [PMID: 38390935 PMCID: PMC10919759 DOI: 10.3892/ijo.2024.5628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Osteosarcoma (OS) is a frequently occurring primary bone tumor, mostly affecting children, adolescents and young adults. Before 1970, surgical resection was the main treatment method for OS, but the clinical results were not promising. Subsequently, the advent of chemotherapy has improved the prognosis of patients with OS. However, there is still a high incidence of metastasis or recurrence, and chemotherapy has several side effects, thus making the 5‑year survival rate markedly low. Recently, chimeric antigen receptor T (CAR‑T) cell therapy represents an alternative immunotherapy approach with significant potential for hematologic malignancies. Nevertheless, the application of CAR‑T cells in the treatment of OS faces numerous challenges. The present review focused on the advances in the development of CAR‑T cells to improve their clinical efficacy, and discussed ways to overcome the difficulties faced by CAR T‑cell therapy for OS.
Collapse
Affiliation(s)
- Tong Yu
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Weibo Jiang
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yang Wang
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Ying Zhou
- Department of Operating Room, The Third Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Jianhang Jiao
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Minfei Wu
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
8
|
Zeng M, Liu C, Gong H, Tang Z, Wen J, Wang S, Xiao S. Therapeutic potential of tyrosine-protein kinase MET in osteosarcoma. Front Mol Biosci 2024; 11:1367331. [PMID: 38596618 PMCID: PMC11002252 DOI: 10.3389/fmolb.2024.1367331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Osteosarcoma, the most prevalent primary bone tumor in children and young adults, can often be successfully treated with standard chemotherapy and surgery when diagnosed at an early stage. However, patients presenting with metastases face significant challenges in achieving a cure. Despite advancements in classical therapies over the past few decades, clinical outcomes for osteosarcoma have not substantially improved. Recently, there has been increased understanding of the biology of osteosarcoma, leading to the identification of new therapeutic targets. One such target is MET, a tyrosine kinase receptor for Hepatocyte Growth Factor (HGF) encoded by the MET gene. In vitro and in vivo studies have demonstrated that the HGF/MET pathway plays a crucial role in cancer growth, invasion, metastasis, and drug resistance across various cancers. Clinical trials targeting this pathway are already underway for lung cancer and hepatocellular carcinoma. Moreover, MET has also been implicated in promoting osteosarcoma progression. This review summarizes 3 decades' worth of research on MET's involvement in osteosarcoma and further explores its potential as a therapeutic target for patients with this disease.
Collapse
Affiliation(s)
- Ming Zeng
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Can Liu
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Haoli Gong
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhongwen Tang
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Sisi Wang
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Sheng Xiao
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
9
|
Al-Ansari N, Samuel SM, Büsselberg D. Unveiling the Protective Role of Melatonin in Osteosarcoma: Current Knowledge and Limitations. Biomolecules 2024; 14:145. [PMID: 38397382 PMCID: PMC10886489 DOI: 10.3390/biom14020145] [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: 12/13/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Melatonin, an endogenous neurohormone produced by the pineal gland, has received increased interest due to its potential anti-cancer properties. Apart from its well-known role in the sleep-wake cycle, extensive scientific evidence has shown its role in various physiological and pathological processes, such as inflammation. Additionally, melatonin has demonstrated promising potential as an anti-cancer agent as its function includes inhibition of tumorigenesis, induction of apoptosis, and regulation of anti-tumor immune response. Although a precise pathophysiological mechanism is yet to be established, several pathways related to the regulation of cell cycle progression, DNA repair mechanisms, and antioxidant activity have been implicated in the anti-neoplastic potential of melatonin. In the current manuscript, we focus on the potential anti-cancer properties of melatonin and its use in treating and managing pediatric osteosarcoma. This aggressive bone tumor primarily affects children and adolescents and is treated mainly by surgical and radio-oncological interventions, which has improved survival rates among affected individuals. Significant disadvantages to these interventions include disease recurrence, therapy-related toxicity, and severe/debilitating side effects that the patients have to endure, significantly affecting their quality of life. Melatonin has therapeutic effects when used for treating osteosarcoma, attributed to its ability to halt cancer cell proliferation and trigger apoptotic cell death, thereby enhancing chemotherapeutic efficacy. Furthermore, the antioxidative function of melatonin alleviates harmful side effects of chemotherapy-induced oxidative damage, aiding in decreasing therapeutic toxicities. The review concisely explains the many mechanisms by which melatonin targets osteosarcoma, as evidenced by significant results from several in vitro and animal models. Nevertheless, if further explored, human trials remain a challenge that could shed light and support its utility as an adjunctive therapeutic modality for treating osteosarcoma.
Collapse
Affiliation(s)
- Nojoud Al-Ansari
- Department of Medical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar;
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
| |
Collapse
|
10
|
Fang F, Yang J, Wang J, Li T, Wang E, Zhang D, Liu X, Zhou C. The role and applications of extracellular vesicles in osteoporosis. Bone Res 2024; 12:4. [PMID: 38263267 PMCID: PMC10806231 DOI: 10.1038/s41413-023-00313-5] [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: 09/14/2023] [Revised: 11/13/2023] [Accepted: 11/28/2023] [Indexed: 01/25/2024] Open
Abstract
Osteoporosis is a widely observed condition characterized by the systemic deterioration of bone mass and microarchitecture, which increases patient susceptibility to fragile fractures. The intricate mechanisms governing bone homeostasis are substantially impacted by extracellular vesicles (EVs), which play crucial roles in both pathological and physiological contexts. EVs derived from various sources exert distinct effects on osteoporosis. Specifically, EVs released by osteoblasts, endothelial cells, myocytes, and mesenchymal stem cells contribute to bone formation due to their unique cargo of proteins, miRNAs, and cytokines. Conversely, EVs secreted by osteoclasts and immune cells promote bone resorption and inhibit bone formation. Furthermore, the use of EVs as therapeutic modalities or biomaterials for diagnosing and managing osteoporosis is promising. Here, we review the current understanding of the impact of EVs on bone homeostasis, including the classification and biogenesis of EVs and the intricate regulatory mechanisms of EVs in osteoporosis. Furthermore, we present an overview of the latest research progress on diagnosing and treating osteoporosis by using EVs. Finally, we discuss the challenges and prospects of translational research on the use of EVs in osteoporosis.
Collapse
Affiliation(s)
- Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jie Yang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jiahe Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tiantian Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Erxiang Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
11
|
Zheng Z, Zhao X, Yuan B, Jiang S, Yan R, Dong X, Yao Q, Liang H. Soy isoflavones induces mitophagy to inhibit the progression of osteosarcoma by blocking the AKT/mTOR signaling pathway. Mol Med 2024; 30:5. [PMID: 38191316 PMCID: PMC10775635 DOI: 10.1186/s10020-024-00778-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: 08/08/2023] [Accepted: 01/02/2024] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Soy isoflavones (SI) is a natural bioactive substance exhibiting beneficial effects on human health. This study aims to elucidate the therapeutic potential of SI in the treatment of osteosarcoma (OS) and to investigate the underlying mechanisms, particularly focusing on mitophagy. METHODS The effects of SI on the proliferation, apoptosis, migration, and invasion of U2OS cells were analyzed. Mitophagy was assessed through multiple parameters: mitochondrial autophagosomes, mitochondrial membrane potential, autophagy-related proteins, reactive oxygen species (ROS), and oxygen consumption rate (OCR). Protein levels related to apoptosis, autophagy, and the AKT/mTOR pathway were analyzed using western blot. The therapeutic efficacy of SI was further identified using a mouse tumor xenograft model. Cell apoptosis and proliferation in tumor xenografts were detected by TUNEL staining and immunohistochemistry (IHC), respectively. RESULTS SI dose-dependently suppressed the viability, colony formation, migration, and invasion of U2OS cells, and enhanced the apoptosis. SI also dose-dependently induced mitophagy in OS cells, evidenced by an increase in autophagosomes and ROS levels, a decrease in mitochondrial membrane potential and OCR, and concomitant changes in autophagy-related proteins. Mdivi-1, an inhibitor of mitophagy, reversed the anti-tumor effects of SI on U2OS cells. In addition, SI blocked the AKT/mTOR pathway in U2OS cells. SC-79, an AKT agonist, reversed the effect of SI on inducing mitophagy. Moreover, SI also promoted cell apoptosis and mitophagy in tumor xenografts in vivo. CONCLUSIONS SI induces mitophagy in OS cells by blocking the AKT/mTOR pathway, contributing to the inhibition of OS.
Collapse
Affiliation(s)
- Ziang Zheng
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Xinghan Zhao
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Bo Yuan
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Shan Jiang
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Rushan Yan
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Xiaowei Dong
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Qijun Yao
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China
| | - Haidong Liang
- Department of Bone and Soft Tissue Repair and Reconstructive Surgery, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian, 116000, Liaoning, China.
| |
Collapse
|
12
|
Tu Z, Li W, Chen Z, Jiang D, Zhou S, Lv S, Cui H. Tumor microenvironment phenotypes and prognostic evaluation tools for osteosarcoma characterized by different prognostic outcomes and immunotherapy responses. J Gene Med 2024; 26:e3572. [PMID: 37525871 DOI: 10.1002/jgm.3572] [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: 05/17/2023] [Revised: 06/28/2023] [Accepted: 07/08/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND The physiological and immunological characteristics of the tumor microenvironment (TME) have a profound impact on the effectiveness of immunotherapy. The present study aimed to define the TME subtype of osteosarcoma according to the signatures representing the global TME of the tumor, as well as create a new prognostic assessment tool to monitor the prognosis, TME activity and immunotherapy response of patients with osteosarcoma. METHODS The enrichment scores of 29 functional gene expression signatures in osteosarcoma samples were calculated by single sample gene set enrichment analysis (ssGSEA). TME classification of osteosarcoma was performed and a prognostic assessment tool was created based on 29 ssGSEA scores to comprehensively correlate them with TME components, immunotherapy efficacy and prognosis of osteosarcoma. RESULTS Three TME subtypes were generated that differed in survival, TME activity and immunotherapeutic response. Four differentially expressed genes between TME subtypes were involved in the development of prognostic assessment tools. The established prognosis assessment tool had strong performance in both training and verification cohorts, could be effectively applied to the survival prediction of samples of different ages, genders and transfer states, and could well distinguish the TME status of different samples. CONCLUSIONS The present study describes three different TME phenotypes in osteosarcoma, provides a risk stratification tool for osteosarcoma prognosis and TME status assessment, and provides additional information for clinical decision-making of immunotherapy.
Collapse
Affiliation(s)
- Zubo Tu
- Orthopedics, Hai'an People's Hospital, Nantong, China
| | - Wang Li
- Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Zhigang Chen
- Orthopedics, Hai'an People's Hospital, Nantong, China
| | - Dong Jiang
- Orthopedics, Hai'an People's Hospital, Nantong, China
| | - Shiran Zhou
- Orthopedics, Hai'an People's Hospital, Nantong, China
| | - Shujun Lv
- Orthopedics, Hai'an People's Hospital, Nantong, China
| | - Haidong Cui
- Orthopedics, Hai'an People's Hospital, Nantong, China
| |
Collapse
|
13
|
Hashemi M, Razzazan M, Bagheri M, Asadi S, Jamali B, Khalafi M, Azimi A, Rad S, Behroozaghdam M, Nabavi N, Rashidi M, Dehkhoda F, Taheriazam A, Entezari M. Versatile function of AMPK signaling in osteosarcoma: An old player with new emerging carcinogenic functions. Pathol Res Pract 2023; 251:154849. [PMID: 37837858 DOI: 10.1016/j.prp.2023.154849] [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: 08/09/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/16/2023]
Abstract
AMP-activated protein kinase (AMPK) signaling has a versatile role in Osteosarcoma (OS), an aggressive bone malignancy with a poor prognosis, particularly in cases that have metastasized or recurred. This review explores the regulatory mechanisms, functional roles, and therapeutic applications of AMPK signaling in OS. It focuses on the molecular activation of AMPK and its interactions with cellular processes like proliferation, apoptosis, and metabolism. The uncertain role of AMPK in cancer is also discussed, highlighting its potential as both a tumor suppressor and a contributor to carcinogenesis. The therapeutic potential of targeting AMPK signaling in OS treatment is examined, including direct and indirect activators like metformin, A-769662, resveratrol, and salicylate. Further research is needed to determine dosing, toxicities, and molecular mechanisms responsible for the anti-osteosarcoma effects of these compounds. This review underscores the complex involvement of AMPK signaling in OS and emphasizes the need for a comprehensive understanding of its molecular mechanisms. By elucidating the role of AMPK in OS, the aim is to pave the way for innovative therapeutic approaches that target this pathway, ultimately improving the prognosis and quality of life for OS patients.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Razzazan
- Medical Student, Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Bagheri
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Behdokht Jamali
- Department of Microbiology and Genetics, Kherad Institute of Higher Education, Bushehr, lran
| | - Maryam Khalafi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics,Faculty of Medicine, Islamic Azad University, Kish International Branch, Kish, Iran
| | - Abolfazl Azimi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics,Faculty of Medicine, Islamic Azad University, Kish International Branch, Kish, Iran
| | - Sepideh Rad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics,Faculty of Medicine, Islamic Azad University, Kish International Branch, Kish, Iran
| | - Mitra Behroozaghdam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Farshid Dehkhoda
- Department of Orthopedics, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
14
|
Nirala BK, Yamamichi T, Petrescu DI, Shafin TN, Yustein JT. Decoding the Impact of Tumor Microenvironment in Osteosarcoma Progression and Metastasis. Cancers (Basel) 2023; 15:5108. [PMID: 37894474 PMCID: PMC10605493 DOI: 10.3390/cancers15205108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Osteosarcoma (OS) is a heterogeneous, highly metastatic bone malignancy in children and adolescents. Despite advancements in multimodal treatment strategies, the prognosis for patients with metastatic or recurrent disease has not improved significantly in the last four decades. OS is a highly heterogeneous tumor; its genetic background and the mechanism of oncogenesis are not well defined. Unfortunately, no effective molecular targeted therapy is currently available for this disease. Understanding osteosarcoma's tumor microenvironment (TME) has recently gained much interest among scientists hoping to provide valuable insights into tumor heterogeneity, progression, metastasis, and the identification of novel therapeutic avenues. Here, we review the current understanding of the TME of OS, including different cellular and noncellular components, their crosstalk with OS tumor cells, and their involvement in tumor progression and metastasis. We also highlight past/current clinical trials targeting the TME of OS for effective therapies and potential future therapeutic strategies with negligible adverse effects.
Collapse
Affiliation(s)
| | | | | | | | - Jason T. Yustein
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA 30322, USA; (B.K.N.); (T.Y.); (D.I.P.); (T.N.S.)
| |
Collapse
|
15
|
Bian J, Liu Y, Zhao X, Meng C, Zhang Y, Duan Y, Wang G. Research progress in the mechanism and treatment of osteosarcoma. Chin Med J (Engl) 2023; 136:2412-2420. [PMID: 37649421 PMCID: PMC10586865 DOI: 10.1097/cm9.0000000000002800] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Indexed: 09/01/2023] Open
Abstract
ABSTRACT Osteosarcoma (OS) is the most common primary malignant bone tumor that more commonly occurs in children and adolescents. The most commonly used treatment for OS is surgery combined with chemotherapy, but the treatment outcomes are typically unsatisfactory. High rates of metastasis and post-treatment recurrence rates are major challenges in the treatment of OS. This underlines the need for studying the in-depth characterization of the pathogenetic mechanisms of OS and development of more effective therapeutic modalities. Previous studies have demonstrated the important role of the bone microenvironment and the regulation of signaling pathways in the occurrence and development of OS. In this review, we discussed the available evidence pertaining to the mechanisms of OS development and identified therapeutic targets for OS. We also summarized the available treatment modalities for OS and identified future priorities for therapeutics research.
Collapse
Affiliation(s)
- Jichao Bian
- Department of Joint and Sports Medicine, The Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, China
| | - Yang Liu
- Department of Pathology, The Second People's Hospital Of Jining, Jining, Shandong 272049, China
| | - Xiaowei Zhao
- Department of Joint and Sports Medicine, The Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, China
| | - Chunyang Meng
- Department of Spine, The Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, China
| | - Yuanmin Zhang
- Department of Joint and Sports Medicine, The Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, China
| | - Yangmiao Duan
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Guodong Wang
- Department of Joint and Sports Medicine, The Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, China
| |
Collapse
|
16
|
An N, Chen Z, Zhao P, Yin W. Extracellular Vesicles in Sepsis: Pathogenic Roles, Organ Damage, and Therapeutic Implications. Int J Med Sci 2023; 20:1722-1731. [PMID: 37928875 PMCID: PMC10620861 DOI: 10.7150/ijms.86832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Despite significant advances in anti-infective treatment and organ function support technology in recent years, the mortality rate of sepsis remains high. In addition to the high costs of sepsis treatment, the increasing consumption of medical resources also aggravates economic pressure and social burden. Extracellular vesicles (EVs) are membrane vesicles released from different types of activated or apoptotic cells to mediate intercellular communication, which can be detected in both human and animal body fluids. A growing body of researches suggest that EVs play an important role in the pathogenesis of sepsis. In this review, we summarize the predominant roles of EVs in various pathological processes during sepsis and its related organ dysfunction.
Collapse
Affiliation(s)
- Ni An
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Zhe Chen
- University College London, London, UK
| | - Peng Zhao
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wen Yin
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
17
|
Urlić I, Jovičić MŠ, Ostojić K, Ivković A. Cellular and Genetic Background of Osteosarcoma. Curr Issues Mol Biol 2023; 45:4344-4358. [PMID: 37232745 DOI: 10.3390/cimb45050276] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
Osteosarcoma describes a tumor of mesenchymal origin with an annual incidence rate of four to five people per million. Even though chemotherapy treatment has shown success in non-metastatic osteosarcoma, metastatic disease still has a low survival rate of 20%. A targeted therapy approach is limited due to high heterogeneity of tumors, and different underlying mutations. In this review, we will summarize new advances obtained by new technologies, such as next generation sequencing and single-cell sequencing. These new techniques have enabled better assessment of cell populations within osteosarcoma, as well as an understanding of the molecular pathogenesis. We also discuss the presence and properties of osteosarcoma stem cells-the cell population within the tumor that is responsible for metastasis, recurrence, and drug resistance.
Collapse
Affiliation(s)
- Inga Urlić
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Marijana Šimić Jovičić
- Department of Paediatric Orthopaedics, Children's Hospital Zagreb, 10000 Zagreb, Croatia
| | - Karla Ostojić
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Alan Ivković
- Department of Orthopaedics and Traumatology, University Hospital Sveti Duh, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Professional Study in Physiotherapy, University of Applied Health Sciences, 10000 Zagreb, Croatia
| |
Collapse
|
18
|
Zhang Y, Wei J, Kong L, Song M, Zhang Y, Xiao X, Cao H, Jin Y. Network pharmacology, molecular docking and bioinformatics reveal the mechanism of Tripterygii Wilfordii against Osteosarcoma. Medicine (Baltimore) 2022; 101:e32389. [PMID: 36595977 PMCID: PMC9803490 DOI: 10.1097/md.0000000000032389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Osteosarcoma (OS) is a malignant bone tumor of mesenchymal origin. Tripterygii Wilfordii (TW) is a traditional Chinese medicine widely used for its anti-inflammatory and immunomodulatory effects. Various components of TW have been shown to have antitumor effects, however, no systematic study has been conducted to prove the anti-OS effects of TW. This study aimed to investigate the effects of TW on OS and its mechanism based on network pharmacology and molecular docking. The web pharmacology section includes the gathering of the active components of TW, the collection of predicted targets of TW and OS-related targets, the analysis of therapeutic targets of TW, the enrichment of gene ontology (GO), and the enrichment of Kyoto Encyclopedia of Genes and Genomes (KEGG). The Veen diagram showed 451 targets for OS treatment in TW. The therapeutic target enrichment analysis results showed that TW treated OS via multiple targets and pathways. TW can affect OS proliferation, apoptosis, migration, infiltration, and angiogenesis through a signaling network formed by hub genes that cascade through numerous signaling pathways. In addition, molecular docking results showed that triptolide, kaempferol, and 5,8-Dihydroxy-7-(4-hydroxy-5-methyl-coumarin-3)-coumarin have relatively high potential to become drugs for patients with OS and improve the 5-year survival rate of patients with OS. Network pharmacology and molecular docking suggest that TW affects the biological behavior of OS through multiple pathways involving multiple targets, such as proliferation, apoptosis, migration, and infiltration. Upregulation of the cellular tumor antigen p53 (TP53) gene and downregulation of peroxisome proliferator-activated receptor gamma (PPARG) and signal transducer and activator of transcription 1-alpha/beta (STAT1) genes can prolong the survival time of patients with OS. Triptolide, kaempferol, and 5,8-Dihydroxy-7-(4-hydroxy-5 methyl-coumarin-3)-coumarin have a relatively high potential to become a treatment for patients with OS and improve 5-year survival of OS patients.
Collapse
Affiliation(s)
- Yafang Zhang
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Junqiang Wei
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Lingwei Kong
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Mingze Song
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Yange Zhang
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Xiangyu Xiao
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Haiying Cao
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Yu Jin
- Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
- *Correspondence: Yu Jin, Department of Traumatology and Orthopaedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, China (e-mail: )
| |
Collapse
|
19
|
The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells. Int J Mol Sci 2022; 23:ijms232416155. [PMID: 36555795 PMCID: PMC9788144 DOI: 10.3390/ijms232416155] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma (OS) is a malignancy that is becoming increasingly common in adolescents. OS stem cells (OSCs) form a dynamic subset of OS cells that are responsible for malignant progression and chemoradiotherapy resistance. The unique properties of OSCs, including self-renewal, multilineage differentiation and metastatic potential, 149 depend closely on their tumor microenvironment. In recent years, the likelihood of its dynamic plasticity has been extensively studied. Importantly, the tumor microenvironment appears to act as the main regulatory component of OS cell plasticity. For these reasons aforementioned, novel strategies for OS treatment focusing on modulating OS cell plasticity and the possibility of modulating the composition of the tumor microenvironment are currently being explored. In this paper, we review recent studies describing the phenomenon of OSCs and factors known to influence phenotypic plasticity. The microenvironment, which can regulate OSC plasticity, has great potential for clinical exploitation and provides different perspectives for drug and treatment design for OS.
Collapse
|
20
|
Matsuzaka Y, Yashiro R. Regulation of Extracellular Vesicle-Mediated Immune Responses against Antigen-Specific Presentation. Vaccines (Basel) 2022; 10:1691. [PMID: 36298556 PMCID: PMC9607341 DOI: 10.3390/vaccines10101691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
Extracellular vesicles (EVs) produced by various immune cells, including B and T cells, macrophages, dendritic cells (DCs), natural killer (NK) cells, and mast cells, mediate intercellular communication and have attracted much attention owing to the novel delivery system of molecules in vivo. DCs are among the most active exosome-secreting cells of the immune system. EVs produced by cancer cells contain cancer antigens; therefore, the development of vaccine therapy that does not require the identification of cancer antigens using cancer-cell-derived EVs may have significant clinical implications. In this review, we summarise the molecular mechanisms underlying EV-based immune responses and their therapeutic effects on tumour vaccination.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan
- Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi 181-8611, Tokyo, Japan
| |
Collapse
|