1
|
Tseng C, Chen CM, Hsieh YH, Lin CY, Chen JW, Hsiao PH, Fong YC, Wang PH, Chen PN, Lin RC. MTA2 knockdown suppresses human osteosarcoma metastasis by inhibiting uPA expression. Aging (Albany NY) 2024; 16:12239-12251. [PMID: 39248711 PMCID: PMC11424574 DOI: 10.18632/aging.206070] [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: 02/23/2024] [Accepted: 07/17/2024] [Indexed: 09/10/2024]
Abstract
The relationship between metastasis-associated protein 2 (MTA2) overexpression and tumor growth and metastasis has been extensively studied in a variety of tumor cells but not in human osteosarcoma cells. This study aims to elucidate the clinical significance, underlying molecular mechanisms, and biological functions of MTA2 in human osteosarcoma in vitro and in vivo. Our results show that MTA2 was elevated in osteosarcoma cell lines and osteosarcoma tissues and was associated with tumor stage and overall survival of osteosarcoma patients. Knockdown of MTA2 inhibited osteosarcoma cell migration and invasion by reducing the expression of urokinase-type plasminogen activator (uPA). Bioinformatic analysis demonstrated that high levels of uPA in human osteosarcoma tissues correlated positively with MTA2 expression. Furthermore, treatment with recombinant human uPA (Rh-uPA) caused significant restoration of OS cell migration and invasion in MTA2 knockdown osteosarcoma cells. We found that ERK1/2 depletion increased the expression of uPA, facilitating osteosarcoma cell migration and invasion. Finally, MTA2 depletion significantly reduced tumor metastasis and the formation of lung nodules in vivo. Overall, our study suggests that MTA2 knockdown suppresses osteosarcoma cell metastasis by decreasing uPA expression via ERK signaling. This finding provides new insight into potential treatment strategies against osteosarcoma metastasis by targeting MTA2.
Collapse
Affiliation(s)
- Chun Tseng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Chien-Min Chen
- Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Department of Leisure Industry Management, National Chin-Yi University of Technology, Taichung, Taiwan
- Department of Biomedical Sciences National Chung Cheng University, Chiayi, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Yu Lin
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
| | - Jian-Wen Chen
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
| | - Pang-Hsuan Hsiao
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Pei-Han Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Renn-Chia Lin
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
2
|
Zhongyu X, Wei X, Hongmei Z, Xiaodong G, Xiaojing Y, Yuanpei L, Li Z, Zhenmin F, Jianda X. Review of pre-metastatic niches induced by osteosarcoma-derived extracellular vesicles in lung metastasis: A potential opportunity for diagnosis and intervention. Biomed Pharmacother 2024; 178:117203. [PMID: 39067163 DOI: 10.1016/j.biopha.2024.117203] [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: 05/30/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Osteosarcoma (OS) has a high propensity for lung metastasis, which is the leading cause of OS-related death and treatment failure. Intercellular communication between OS cells and distant lung host cells is required for the successful lung metastasis of OS cells to the lung. Before OS cells infiltrate the lung, in situ OS cells secrete extracellular vesicles (EVs) that act as mediators of cell-to-cell communication. In recent years, EVs have been confirmed to act as bridges and key drivers between in situ tumors and metastatic lesions by regulating the formation of a pre-metastatic niche (PMN), defined as a microenvironment suitable for disseminated tumor cell engraftment and colonization, in distant target organs. This review summarizes the current knowledge about the underlying mechanisms of PMN formation induced by OS-derived EVs and the potential roles of EVs as targets or drug carriers in regulating PMN formation in the lung. We also provide an overview of their potential EV-based therapeutic strategies for hindering PMN formation in the context of OS lung metastasis.
Collapse
Affiliation(s)
- Xia Zhongyu
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Xu Wei
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, Jiangsu 224051, China
| | - Zhang Hongmei
- School of Mechanical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Ge Xiaodong
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, Jiangsu 224051, China
| | - Yan Xiaojing
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Lian Yuanpei
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Zhu Li
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Fan Zhenmin
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou Jiangsu, China.
| | - Xu Jianda
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China.
| |
Collapse
|
3
|
Pankova V, Krasny L, Kerrison W, Tam YB, Chadha M, Burns J, Wilding CP, Chen L, Chowdhury A, Perkins E, Lee AT, Howell L, Guljar N, Sisley K, Fisher C, Chudasama P, Thway K, Jones RL, Huang PH. Clinical Implications and Molecular Features of Extracellular Matrix Networks in Soft Tissue Sarcomas. Clin Cancer Res 2024; 30:3229-3242. [PMID: 38810090 PMCID: PMC11292195 DOI: 10.1158/1078-0432.ccr-23-3960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/25/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE The landscape of extracellular matrix (ECM) alterations in soft tissue sarcomas (STS) remains poorly characterized. We aimed to investigate the tumor ECM and adhesion signaling networks present in STS and their clinical implications. EXPERIMENTAL DESIGN Proteomic and clinical data from 321 patients across 11 histological subtypes were analyzed to define ECM and integrin adhesion networks. Subgroup analysis was performed in leiomyosarcomas (LMS), dedifferentiated liposarcomas (DDLPS), and undifferentiated pleomorphic sarcomas (UPS). RESULTS This analysis defined subtype-specific ECM profiles including enrichment of basement membrane proteins in LMS and ECM proteases in UPS. Across the cohort, we identified three distinct coregulated ECM networks which are associated with tumor malignancy grade and histological subtype. Comparative analysis of LMS cell line and patient proteomic data identified the lymphocyte cytosolic protein 1 cytoskeletal protein as a prognostic factor in LMS. Characterization of ECM network events in DDLPS revealed three subtypes with distinct oncogenic signaling pathways and survival outcomes. Evaluation of the DDLPS subtype with the poorest prognosis nominates ECM remodeling proteins as candidate antistromal therapeutic targets. Finally, we define a proteoglycan signature that is an independent prognostic factor for overall survival in DDLPS and UPS. CONCLUSIONS STS comprise heterogeneous ECM signaling networks and matrix-specific features that have utility for risk stratification and therapy selection, which could in future guide precision medicine in these rare cancers.
Collapse
Affiliation(s)
- Valeriya Pankova
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Lukas Krasny
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - William Kerrison
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Yuen B. Tam
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Madhumeeta Chadha
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Jessica Burns
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Christopher P. Wilding
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Liang Chen
- Precision Sarcoma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- National Center for Tumor Diseases, Heidelberg, Germany.
| | - Avirup Chowdhury
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Emma Perkins
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | | | - Louise Howell
- Light Microscopy Facility, The Institute of Cancer Research, London, United Kingdom.
| | - Nafia Guljar
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| | - Karen Sisley
- Division of Clinical Medicine, The Medical School, University of Sheffield, Sheffield, United Kingdom.
| | - Cyril Fisher
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom.
| | - Priya Chudasama
- Precision Sarcoma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- National Center for Tumor Diseases, Heidelberg, Germany.
| | - Khin Thway
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
- The Royal Marsden NHS Foundation Trust, London, United Kingdom.
| | - Robin L. Jones
- The Royal Marsden NHS Foundation Trust, London, United Kingdom.
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
4
|
Fu Z, Sun G, Li J, Yu H. Identification of hub genes related to metastasis and prognosis of osteosarcoma and establishment of a prognostic model with bioinformatic methods. Medicine (Baltimore) 2024; 103:e38470. [PMID: 38847690 PMCID: PMC11155596 DOI: 10.1097/md.0000000000038470] [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: 07/30/2023] [Accepted: 05/15/2024] [Indexed: 06/10/2024] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor occurring in children and adolescents. Improvements in our understanding of the OS pathogenesis and metastatic mechanism on the molecular level might lead to notable advances in the treatment and prognosis of OS. Biomarkers related to OS metastasis and prognosis were analyzed and identified, and a prognostic model was established through the integration of bioinformatics tools and datasets in multiple databases. 2 OS datasets were downloaded from the Gene Expression Omnibus database for data consolidation, standardization, batch effect correction, and identification of differentially expressed genes (DEGs); following that, gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the DEGs; the STRING database was subsequently used for protein-protein interaction (PPI) network construction and identification of hub genes; hub gene expression was validated, and survival analysis was conducted through the employment of the TARGET database; finally, a prognostic model was established and evaluated subsequent to the screening of survival-related genes. A total of 701 DEGs were identified; by gene ontology and KEGG pathway enrichment analyses, the overlapping DEGs were enriched for 249 biological process terms, 13 cellular component terms, 35 molecular function terms, and 4 KEGG pathways; 13 hub genes were selected from the PPI network; 6 survival-related genes were identified by the survival analysis; the prognostic model suggested that 4 genes were strongly associated with the prognosis of OS. DEGs related to OS metastasis and survival were identified through bioinformatics analysis, and hub genes were further selected to establish an ideal prognostic model for OS patients. On this basis, 4 protective genes including TPM1, TPM2, TPM3, and TPM4 were yielded by the prognostic model.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
- Department of Orthopedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Guofeng Sun
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| | - Jingtian Li
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| | - Hongjian Yu
- Department of Orthopedics, Binzhou People’s Hospital, Binzhou,China
| |
Collapse
|
5
|
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
|
6
|
Chen CC, Benavente CA. Exploring the Impact of Exosomal Cargos on Osteosarcoma Progression: Insights into Therapeutic Potential. Int J Mol Sci 2024; 25:568. [PMID: 38203737 PMCID: PMC10779183 DOI: 10.3390/ijms25010568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor with high metastasis. Poor prognosis highlights a clinical need for novel therapeutic strategies. Exosomes, also known as extracellular vesicles, have been identified as essential players in the modulation of cancer. Recent studies have suggested that OS-derived exosomes can drive pro-tumorigenic or anti-tumorigenic phenotypes by transferring specific cargos, including proteins, nucleic acids, and metabolites, to neighboring cells, significantly impacting the regulation of cellular processes. This review discusses the advancement of exosomes and their cargos in OS. We examine how these exosomes contribute to the modulation of cellular phenotypes associated with tumor progression and metastasis. Furthermore, we explore the potential of exosomes as valuable biomarkers for diagnostics and prognostic purposes and their role in shaping innovative therapeutic strategies in OS treatment development.
Collapse
Affiliation(s)
- Claire C. Chen
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA;
| | - Claudia A. Benavente
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA;
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
| |
Collapse
|
7
|
Liao Y, Yi Q, He J, Huang D, Xiong J, Sun W, Sun W. Extracellular vesicles in tumorigenesis, metastasis, chemotherapy resistance and intercellular communication in osteosarcoma. Bioengineered 2023; 14:113-128. [PMID: 37377390 DOI: 10.1080/21655979.2022.2161711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 06/29/2023] Open
Abstract
HIGHLIGHTS Extracellular vehicles play crucial function in osteosarcoma tumorigenesis.Extracellular vehicles mediated the intercellular communication of osteosarcoma cells with other types cells in tumor microenvironment.Extracellular vehicles have potential utility in osteosarcoma diagnosis and treatment.
Collapse
Affiliation(s)
- Yi Liao
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University, Chongqing, Chongqing, China
| | - Qian Yi
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, China
- The Central Laboratory, Affiliated Hospital of Putian University, Putian, Fujian, China
| | - Jinglong He
- Department of Orthopaedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
| | - Dixi Huang
- Guangzhou Medical University, Guangzhou, China
| | - Jianyi Xiong
- Department of Orthopaedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
| | - Wei Sun
- Department of Orthopaedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
| | - Weichao Sun
- Department of Orthopaedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
| |
Collapse
|
8
|
Jiang M, Jike Y, Liu K, Gan F, Zhang K, Xie M, Zhang J, Chen C, Zou X, Jiang X, Dai Y, Chen W, Qiu Y, Bo Z. Exosome-mediated miR-144-3p promotes ferroptosis to inhibit osteosarcoma proliferation, migration, and invasion through regulating ZEB1. Mol Cancer 2023; 22:113. [PMID: 37461104 PMCID: PMC10351131 DOI: 10.1186/s12943-023-01804-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most prevalent orthopedic malignancy with a dismal prognosis. The high iron absorption rate in OS cells of patients suggests that ferroptosis may be related to the progression of OS, but its potential molecular regulatory role is still unclear. Based on the ability to couple with exosomes for targeted delivery of signals, exosome-derived micro ribonucleic acids (miRNAs) can potentially serve as diagnostic biomarkers for OS. METHODS We identified ferroptosis-related miRNAs and messenger ribonucleic acids(mRNAs) in OS using bioinformatics analysis and performed survival analysis. Then we measured miRNA expression levels through exosome microarray sequencing, and used RT-qPCR and IHC to verify the expression level of miR-144-3p and ZEB1. Stable gene expression cell lines were fabricated for in vitro experiments. Cell viability, migration and invasion were determined by CCK-8 and transwell experiment. Use the corresponding reagent kit to detect GSH/GSSG ratio, Fe2+ level, MDA level and ROS level, and measure the expression levels of GPX4, ACSL4 and xCT through RT-qPCR and WB. We also constructed nude mice model for in vivo experiments. Finally, the stability of the miRNA/mRNA axis was verified through functional rescue experiments. RESULTS Low expression of miR-144-3p and high expression of ZEB1 in OS cell lines and tissues was observed. Overexpression of miR-144-3p can promote ferroptosis, reduce the survival ability of OS cells, and prevent the progression of OS. In addition, overexpression of miR-144-3p can downregulate the expression of ZEB1 in cell lines and nude mice. Knockdown of miR-144-3p has the opposite effect. The functional rescue experiment validated that miR-144-3p can regulate downstream ZEB1, and participates in the occurrence and development of OS by interfering with redox homeostasis and iron metabolism. CONCLUSIONS MiR-144-3p can induce the occurrence of ferroptosis by negatively regulating the expression of ZEB1, thereby inhibiting the proliferation, migration, and invasion of OS cells.
Collapse
Affiliation(s)
- Mingyang Jiang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yiji Jike
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Kaicheng Liu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fu Gan
- Department of Urology Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Ke Zhang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mingjing Xie
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Junlei Zhang
- Department of Sports Medicine, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Chuanliang Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaochong Zou
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaohong Jiang
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongheng Dai
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Weikui Chen
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yue Qiu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhandong Bo
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
9
|
Agnoletto C, Pignochino Y, Caruso C, Garofalo C. Exosome-Based Liquid Biopsy Approaches in Bone and Soft Tissue Sarcomas: Review of the Literature, Prospectives, and Hopes for Clinical Application. Int J Mol Sci 2023; 24:ijms24065159. [PMID: 36982236 PMCID: PMC10048895 DOI: 10.3390/ijms24065159] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
The knowledge of exosome impact on sarcoma development and progression has been implemented in preclinical studies thanks to technological advances in exosome isolation. Moreover, the clinical relevance of liquid biopsy is well established in early diagnosis, prognosis prediction, tumor burden assessment, therapeutic responsiveness, and recurrence monitoring of tumors. In this review, we aimed to comprehensively summarize the existing literature pointing out the clinical relevance of detecting exosomes in liquid biopsy from sarcoma patients. Presently, the clinical utility of liquid biopsy based on exosomes in patients affected by sarcoma is under debate. The present manuscript collects evidence on the clinical impact of exosome detection in circulation of sarcoma patients. The majority of these data are not conclusive and the relevance of liquid biopsy-based approaches in some types of sarcoma is still insufficient. Nevertheless, the utility of circulating exosomes in precision medicine clearly emerged and further validation in larger and homogeneous cohorts of sarcoma patients is clearly needed, requiring collaborative projects between clinicians and translational researchers for these rare cancers.
Collapse
Affiliation(s)
| | - Ymera Pignochino
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy
- Candiolo Cancer Instute, FPO-IRCCS, 10060 Torino, Italy
| | - Chiara Caruso
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - Cecilia Garofalo
- Advanced Translational Research Laboratory, Immunology and Molecular Oncology Diagnostic Unit, Veneto Institute of Oncology IOV-IRCCS, 35127 Padua, Italy
| |
Collapse
|
10
|
The Roles of Exosomes in Metastasis of Sarcoma: From Biomarkers to Therapeutic Targets. Biomolecules 2023; 13:biom13030456. [PMID: 36979391 PMCID: PMC10046038 DOI: 10.3390/biom13030456] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Sarcoma is a heterogeneous group of mesenchymal neoplasms with a high rate of lung metastasis. The cellular mechanisms responsible for sarcoma metastasis remain poorly understood. Furthermore, there are limited efficacious therapeutic strategies for treating metastatic sarcoma. Improved diagnostic and therapeutic modalities are of increasing importance for the treatment of sarcoma due to their high mortality in the advanced stages of the disease. Recent evidence demonstrates that the exosome, a type of extracellular vesicle released by virtually all cells in the body, is an important facilitator of intercellular communication between the cells and the surrounding environment. The exosome is gaining significant attention among the medical research community, but there is little knowledge about how the exosome affects sarcoma metastasis. In this review, we summarize the multifaceted roles of sarcoma-derived exosomes in promoting the process of metastasis via the formation of pre-metastatic niche (PMN), the regulation of immunity, angiogenesis, vascular permeability, and the migration of sarcoma cells. We also highlight the potential of exosomes as innovative diagnostic and prognostic biomarkers as well as therapeutic targets in sarcoma metastasis.
Collapse
|
11
|
Akhtar MR, Mondal MNI, Rana HK. Bioinformatics approach to identify the impacts of microgravity on the development of bone and joint diseases. INFORMATICS IN MEDICINE UNLOCKED 2023. [DOI: 10.1016/j.imu.2023.101211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023] Open
|
12
|
Asanuma K, Nakamura T, Okamoto T, Hagi T, Kita K, Nakamura K, Matsuyama Y, Yoshida K, Asanuma Y, Sudo A. Do coagulation or fibrinolysis reflect the disease condition in patients with soft tissue sarcoma? BMC Cancer 2022; 22:1075. [PMID: 36258189 PMCID: PMC9580209 DOI: 10.1186/s12885-022-10106-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coagulation and fibrinolysis are distinct processes that are highly correlated. Cells control coagulation and fibrinolysis by expression of tissue factor and urokinase-type plasminogen activator receptor on their surface. Tumor cells express these proteins, adjust their microenvironment and induce tumor exacerbation. We hypothesized that the expression of plasma markers for coagulation and fibrinolysis in patients with soft tissue sarcomas (STSs) was dependent on the level of tumor malignancy. To elucidate which markers are predictive of recurrence, metastasis and prognosis, coagulation or fibrinolysis, we analyzed the correlation between plasma levels of thrombin-antithrombin III complex (TAT), soluble fibrin (SF), plasmin-α2 plasmin inhibitor complex (PIC), D-dimer (DD) and clinical parameters in patients with STSs. METHODS TAT, SF, PIC or DD were measured in pre-treatment blood samples from 64 patients with primary STSs and analyzed with clinicopathological parameters, and 5-year recurrence free survival (RFS), 5-year metastasis free survival (MFS) and 5-year overall survival (OS) were evaluated. RESULTS The metastasis group had significantly higher DD (p = 0.0394), PIC (p = 0.00532) and SF (p = 0.00249) concentrations than the group without metastasis. The group that died of disease showed significantly higher DD (p = 0.00105), PIC (p = 0.000542), SF (p = 0.000126) and TAT (p = 0.0373) than surviving patients. By dividing the patients into low and high groups, the group with high DD, PIC, SF and TAT showed significantly lower 5-year MFS and 5-year OS than the corresponding low group. Furthermore, in multivariate COX proportional hazard analysis of continuous variables for 5-year MFS, only PIC was found to be a significant factor (HR: 2.14). CONCLUSION Fibrinolysis was better than coagulation at reflecting the disease condition of patients with STS. Notably, PIC levels ≥ 1.1 can not only predict the risk of metastasis and poor prognosis, but also increasing PIC levels correspond to further increases in risks of metastasis and poor prognosis.
Collapse
Affiliation(s)
- Kunihiro Asanuma
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan.
| | - Tomoki Nakamura
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| | - Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, Izumo, Shimane, Japan
| | - Tomohito Hagi
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| | - Kouji Kita
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| | - Koichi Nakamura
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| | - Yumi Matsuyama
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| | - Keisuke Yoshida
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| | - Yumiko Asanuma
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| | - Akihiro Sudo
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, 514-8507, Tsu City, Mie, Japan
| |
Collapse
|
13
|
Wu SC, Kim A, Gu Y, Martinez DI, Zocchi L, Chen CC, Lopez J, Salcido K, Singh S, Wu J, Nael A, Benavente CA. UHRF1 overexpression promotes osteosarcoma metastasis through altered exosome production and AMPK/SEMA3E suppression. Oncogenesis 2022; 11:51. [PMID: 36068209 PMCID: PMC9448786 DOI: 10.1038/s41389-022-00430-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Loss-of-function mutations at the retinoblastoma (RB1) gene are associated with increased mortality, metastasis, and poor therapeutic outcome in several cancers, including osteosarcoma. However, the mechanism(s) through which RB1 loss worsens clinical outcome remains understudied. Ubiquitin-like with PHD and Ring Finger domains 1 (UHRF1) has been identified as a critical downstream effector of the RB/E2F signaling pathway that is overexpressed in various cancers. Here, we determined the role and regulatory mechanisms of UHRF1 in rendering osteosarcoma cells more aggressive. Higher UHRF1 expression correlated with malignancy in osteosarcoma cell lines, clinical samples, and genetically engineered mouse models. Gain- and loss-of-function assays revealed that UHRF1 has cell-intrinsic and extrinsic functions promoting cell proliferation, migration, invasion, angiogenesis, and metastasis. UHRF1 overexpression induced angiogenesis by suppressing AMPK activation and Semaphorin 3E (SEMA3E) expression. Further, UHRF1-mediated migration and metastasis resulted, at least in part, through altered expression of extracellular vesicles and their cargo, including urokinase-type plasminogen activator (uPA). Novel osteosarcoma genetically engineered mouse models confirmed that knocking out Uhrf1 considerably decreased metastasis and reversed the poorer survival associated with Rb1 loss. This presents a new mechanistic insight into RB1 loss-associated poor prognosis and novel oncogenic roles of UHRF1 in the regulation of angiogenesis and exosome secretion, both critical for osteosarcoma metastasis. This provides substantial support for targeting UHRF1 or its downstream effectors as novel therapeutic options to improve current treatment for osteosarcoma.
Collapse
Affiliation(s)
- Stephanie C Wu
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Ahhyun Kim
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Yijun Gu
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Daniel I Martinez
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Loredana Zocchi
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Claire C Chen
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Jocelyne Lopez
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Kelsey Salcido
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Sarah Singh
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, CA, 92697, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, 92697, USA
| | - Ali Nael
- Department of Pathology, University of California, Irvine, CA, 92697, USA
- Department of Pathology, Children's Hospital of Orange County, Orange, CA, 92868, USA
| | - Claudia A Benavente
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA.
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
14
|
Ye H, Hu X, Wen Y, Tu C, Hornicek F, Duan Z, Min L. Exosomes in the tumor microenvironment of sarcoma: from biological functions to clinical applications. J Nanobiotechnology 2022; 20:403. [PMID: 36064358 PMCID: PMC9446729 DOI: 10.1186/s12951-022-01609-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
The current diagnosis and treatment of sarcoma continue to show limited timeliness and efficacy. In order to enable the early detection and management of sarcoma, increasing attentions have been given to the tumor microenvironment (TME). TME is a dynamic network composed of multiple cells, extracellular matrix, vasculature, and exosomes. Exosomes are nano-sized extracellular vesicles derived from various cells in the TME. The major function of exosomes is to promote cancer progress and metastasis through mediating bidirectional cellular communications between sarcoma cells and TME cells. Due to the content specificity, cell tropism, and bioavailability, exosomes have been regarded as promising diagnostic and prognostic biomarkers, and therapeutic vehicles for sarcoma. This review summarizes recent studies on the roles of exosomes in TME of sarcoma, and explores the emerging clinical applications.
Collapse
Affiliation(s)
- Huali Ye
- West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xin Hu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wen
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Chongqi Tu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Li Min
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Zou Z, Sun W, Xu Y, Liu W, Zhong J, Lin X, Chen Y. Application of Multi-Omics Approach in Sarcomas: A Tool for Studying Mechanism, Biomarkers, and Therapeutic Targets. Front Oncol 2022; 12:946022. [PMID: 35875106 PMCID: PMC9304858 DOI: 10.3389/fonc.2022.946022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/16/2022] [Indexed: 12/18/2022] Open
Abstract
Sarcomas are rare, heterogeneous mesenchymal neoplasms with various subtypes, each exhibiting unique genetic characteristics. Although studies have been conducted to improve the treatment for sarcomas, the specific development from normal somatic cells to sarcoma cells is still unclear and needs further research. The diagnosis of sarcomas depends heavily on the pathological examination, which is yet a difficult work and requires expert analysis. Advanced treatment like precise medicine optimizes the efficacy of treatment and the prognosis of sarcoma patients, yet, in sarcomas, more studies should be done to put such methods in clinical practice. The revolution of advanced technology has pushed the multi-omics approach to the front, and more could be learnt in sarcomas with such methods. Multi-omics combines the character of each omics techniques, analyzes the mechanism of tumor cells from different levels, which makes up for the shortage of single-omics, and gives us an integrated picture of bioactivities inside tumor cells. Multi-omics research of sarcomas has reached appreciable progress in recent years, leading to a better understanding of the mutation, proliferation, and metastasis of sarcomas. With the help of multi-omics approach, novel biomarkers were found, with promising effects in improving the process of diagnosis, prognosis anticipation, and treatment decision. By analyzing large amounts of biological features, subtype clustering could be done in a better precision, which may be useful in the clinical procedure. In this review, we summarized recent discoveries using multi-omics approach in sarcomas, discussed their merits and challenges, and concluded with future perspectives of the sarcoma research.
Collapse
Affiliation(s)
- Zijian Zou
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Sun
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Xu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wanlin Liu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingqin Zhong
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyi Lin
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong Chen
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Chu SC, Hsieh YS, Hsu LS, Lin CY, Lai YA, Chen PN. Cinnamaldehyde decreases the invasion and u-PA expression of osteosarcoma by down-regulating the FAK signalling pathway. Food Funct 2022; 13:6574-6582. [PMID: 35678522 DOI: 10.1039/d2fo00634k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer metastasis is the major cause of the high mortality risk of patients with osteosarcoma. Cinnamaldehyde has been shown to exhibit multiple tumour-suppressing activities, but its role in human osteosarcoma is not yet completely defined. In this study, the antimetastatic effect of cinnamaldehyde on highly metastatic human osteosarcoma cells was observed in vitro and in vivo using Saos-2 and 143B cells. Cinnamaldehyde reduced the activity and protein level of urokinase-type plasminogen activator (u-PA) and suppressed the invasion ability of osteosarcoma cells by inhibiting the phosphorylation of focal adhesion kinase. In addition, cinnamaldehyde reduced cell movement, cell-matrix adhesion, and the expression of the mesenchymal markers of epithelial-to-mesenchymal transition, namely, fibronectin and N-cadherin. Importantly, the oral administration of cinnamaldehyde remarkably suppressed the pulmonary metastasis of osteosarcoma in mice. Results indicated that cinnamaldehyde has therapeutic potential for inhibiting osteosarcoma metastasis.
Collapse
Affiliation(s)
- Shu-Chen Chu
- Institute and Department of Food Science, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Yih-Shou Hsieh
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Li-Sung Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | - Chin-Yin Lin
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | - Yi-An Lai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan. .,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
17
|
Ran Q, Xu D, Wang Q, Wang D. Hypermethylation of the Promoter Region of miR-23 Enhances the Metastasis and Proliferation of Multiple Myeloma Cells via the Aberrant Expression of uPA. Front Oncol 2022; 12:835299. [PMID: 35707350 PMCID: PMC9189361 DOI: 10.3389/fonc.2022.835299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/02/2022] [Indexed: 11/24/2022] Open
Abstract
Multiple myeloma has a long course, with no obvious symptoms in the early stages. However, advanced stages are characterized by injury to the bone system and represent a severe threat to human health. The results of the present work indicate that the hypermethylation of miR-23 promoter mediates the aberrant expression of uPA/PLAU (urokinase plasminogen activator, uPA) in multiple myeloma cells. miR-23, a microRNA that potentially targets uPA’s 3’UTR, was predicted by the online tool miRDB. The endogenous expressions of uPA and miR-23 are related to disease severity in human patients, and the expression of miR-23 is negatively related to uPA expression. The hypermethylation of the promoter region of miR-23 is a promising mechanism to explain the low level of miR-23 or aberrant uPA expression associated with disease severity. Overexpression of miR-23 inhibited the expression of uPA by targeting the 3’UTR of uPA, not only in MM cell lines, but also in patient-derived cell lines. Overexpression of miR-23 also inhibited in vitro and in vivo invasion of MM cells in a nude mouse model. The results therefore extend our knowledge about uPA in MM and may assist in the development of more effective therapeutic strategies for MM treatment.
Collapse
Affiliation(s)
- Qijie Ran
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
- *Correspondence: Qijie Ran, ; Dongsheng Wang,
| | - Dehong Xu
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
| | - Qi Wang
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
| | - Dongsheng Wang
- Department of Neurosurgery, The Fifth People’s Hospital of Dalian, Dalian, China
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian City, China
- *Correspondence: Qijie Ran, ; Dongsheng Wang,
| |
Collapse
|
18
|
Adib A, Sahu R, Mohta S, Pollock RE, Casadei L. Cancer-Derived Extracellular Vesicles: Their Role in Sarcoma. Life (Basel) 2022; 12:life12040481. [PMID: 35454972 PMCID: PMC9029613 DOI: 10.3390/life12040481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Soft tissue sarcomas (STS) are rare malignancies with limited responses to anticancer therapy. Extracellular vesicles (EVs) are a heterogeneous group of bi-lipid layer sacs secreted by cells into extracellular space. Investigations of tumor-derived EVs have revealed their functional capabilities, including cell-to-cell communication and their impact on tumorigenesis, progression, and metastasis; however information on the roles of EVs in sarcoma is currently limited. In this review we investigate the role of various EV cargos in sarcoma and the mechanisms by which those cargos can affect the recipient cell phenotype and the aggressivity of the tumor itself. The study of EVs in sarcoma may help establish novel therapeutic approaches that target specific sarcoma subtypes or biologies, thereby improving sarcoma therapeutics in the future.
Collapse
Affiliation(s)
- Anita Adib
- The James Cancer Hospital Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; (S.M.); (L.C.)
- Correspondence: (A.A.); (R.S.)
| | - Ruhi Sahu
- The James Cancer Hospital Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; (S.M.); (L.C.)
- Correspondence: (A.A.); (R.S.)
| | - Shivangi Mohta
- The James Cancer Hospital Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; (S.M.); (L.C.)
| | - Raphael Etomar Pollock
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, OH 43210, USA;
| | - Lucia Casadei
- The James Cancer Hospital Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; (S.M.); (L.C.)
| |
Collapse
|
19
|
Abstract
Osteosarcoma is the most common primary bone malignancy in adolescents. Its high propensity to metastasize is the leading cause for treatment failure and poor prognosis. Although the research of osteosarcoma has greatly expanded in the past decades, the knowledge and new therapy strategies targeting metastatic progression remain sparse. The prognosis of patients with metastasis is still unsatisfactory. There is resonating urgency for a thorough and deeper understanding of molecular mechanisms underlying osteosarcoma to develop innovative therapies targeting metastasis. Toward the goal of elaborating the characteristics and biological behavior of metastatic osteosarcoma, it is essential to combine the diverse investigations that are performed at molecular, cellular, and animal levels from basic research to clinical translation spanning chemical, physical sciences, and biology. This review focuses on the metastatic process, regulatory networks involving key molecules and signaling pathways, the role of microenvironment, osteoclast, angiogenesis, metabolism, immunity, and noncoding RNAs in osteosarcoma metastasis. The aim of this review is to provide an overview of current research advances, with the hope to discovery druggable targets and promising therapy strategies for osteosarcoma metastasis and thus to overcome this clinical impasse.
Collapse
Affiliation(s)
- Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
De Martino V, Rossi M, Battafarano G, Pepe J, Minisola S, Del Fattore A. Extracellular Vesicles in Osteosarcoma: Antagonists or Therapeutic Agents? Int J Mol Sci 2021; 22:12586. [PMID: 34830463 PMCID: PMC8619425 DOI: 10.3390/ijms222212586] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma (OS) is a skeletal tumor affecting mainly children and adolescents. The presence of distance metastasis is frequent and it is localized preferentially to the lung, representing the main reason for death among patients. The therapeutic approaches are based on surgery and chemotherapeutics. However, the drug resistance and the side effects associated with the chemotherapy require the identification of new therapeutic approaches. The understanding of the complex biological scenario of the osteosarcoma will open the way for the identification of new targets for its treatment. Recently, a great interest of scientific community is for extracellular vesicles (EVs), that are released in the tumor microenvironment and are important regulators of tumor proliferation and the metastatic process. At the same time, circulating extracellular vesicles can be exploited as diagnostic and prognostic biomarkers, and they can be loaded with drugs as a new therapeutic approach for osteosarcoma patients. Thus, the characterization of OS-related EVs could represent a way to convert these vesicles from antagonists for human health into therapeutic and/or diagnostic agents.
Collapse
Affiliation(s)
- Viviana De Martino
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy; (V.D.M.); (J.P.); (S.M.)
| | - Michela Rossi
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.R.); (G.B.)
| | - Giulia Battafarano
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.R.); (G.B.)
| | - Jessica Pepe
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy; (V.D.M.); (J.P.); (S.M.)
| | - Salvatore Minisola
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy; (V.D.M.); (J.P.); (S.M.)
| | - Andrea Del Fattore
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.R.); (G.B.)
| |
Collapse
|
21
|
Yang L, Huang X, Guo H, Wang L, Yang W, Wu W, Jing D, Shao Z. Exosomes as Efficient Nanocarriers in Osteosarcoma: Biological Functions and Potential Clinical Applications. Front Cell Dev Biol 2021; 9:737314. [PMID: 34712664 PMCID: PMC8546119 DOI: 10.3389/fcell.2021.737314] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is the most common bone tumor affecting both adolescents and children. Although localized osteosarcoma has an overall survival of >70% in the clinic, metastatic, refractory, and recurrent osteosarcoma have poorer survival rates. Exosomes are extracellular vesicles released by cells and originally thought to be a way for cells to discard unwanted products. Currently, exosomes have been reported to be involved in intercellular cross-talk and induce changes in cellular behavior by transferring cargoes (proteins, DNA, RNA, and lipids) between cells. Exosomes regulate osteosarcoma progression, and processes such as tumorigenesis, proliferation, metastasis, angiogenesis, immune evasion, and drug resistance. Increasing evidences shows that exosomes have significant potential in promoting osteosarcoma progression and development. In this review, we describe the current research status of exosomes in osteosarcoma, focusing on the biological functions of osteosarcoma exosomes as well as their application in osteosarcoma as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Lingkai Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoyu Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lutong Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbo Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Doudou Jing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Cappariello A, Rucci N. Extracellular Vesicles in Bone Tumors: How to Seed in the Surroundings Molecular Information for Malignant Transformation and Progression. Front Oncol 2021; 11:722922. [PMID: 34616676 PMCID: PMC8488258 DOI: 10.3389/fonc.2021.722922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/26/2021] [Indexed: 12/19/2022] Open
Abstract
Bone is a very dynamic tissue hosting different cell types whose functions are regulated by a plethora of membrane-bound and soluble molecules. Intercellular communication was recently demonstrated to be also sustained by the exchange of extracellular vesicles (EVs). These are cell-derived nanosized structures shuttling biologically active molecules, such as nucleic acids and proteins. The bone microenvironment is a preferential site of primary and metastatic tumors, in which cancer cells find a fertile soil to “seed and blossom”. Nowadays, many oncogenic processes are recognized to be sustained by EVs. For example, EVs can directly fuel the vicious cycle in the bone/bone marrow microenvironment. EVs create a favourable environment for tumor growth by affecting osteoblasts, osteoclasts, osteocytes, adipocytes, leukocytes, and endothelial cells. At the same time other crucial tumor-mediated events, such as the premetastatic niche formation, tumor cell dormancy, as well as drug resistance, have been described to be fostered by tumor-derived EVs. In this review, we will discuss the main body of literature describing how the cancer cells use the EVs for their growth into the bone and for educating the bone microenvironment to host metastases.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
23
|
Zhang XB, Zhang RH, Su X, Qi J, Hu YC, Shi JT, Zhang K, Wang KP, Zhou HY. Exosomes in osteosarcoma research and preclinical practice. Am J Transl Res 2021; 13:882-897. [PMID: 33841628 PMCID: PMC8014357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/15/2020] [Indexed: 06/12/2023]
Abstract
Osteosarcoma (OS) is a rare soft-tissue malignant tumor with high lung metastasis and mortality rates. Preoperative chemotherapy, surgical resection of the lesion and postoperative chemotherapy are still the main treatments for osteosarcoma. The prognosis, however, is poor for patients with nonresectable, primary metastatic or relapsed disease. Recent studies have shown that targeted therapy for OS based on the characteristics of exosomes is very attractive. Exosomes are nanosized extracellular vesicles (EVs) that participate in cell-to-cell communication by transporting biologically active cargo molecules, causing changes in OS cell function and playing important roles in OS disease progression. With the characteristics of secretory cells, exosomes transport cargo (e.g., microRNAs) that can be used to detect the progress of a disease and can serve as markers and/or therapeutic targets for clinical diagnosis of OS. In this review, the roles of exosomes in OS pathogenesis, invasion, metastasis, drug resistance, diagnosis and treatment are summarized. In addition, this article elaborates a series of challenges to overcome before exosomes are applied in clinical practice and provides suggestions based on current evidence for the direction of future research.
Collapse
Affiliation(s)
- Xiao-Bo Zhang
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu ProvinceLanzhou 730000, China
| | - Rui-Hao Zhang
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu ProvinceLanzhou 730000, China
| | - Xin Su
- Department of Cardiology, Gansu Province People’s HospitalLanzhou 730000, China
| | - Jin Qi
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu ProvinceLanzhou 730000, China
| | - Yi-Cun Hu
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu ProvinceLanzhou 730000, China
| | - Jin-Tao Shi
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu ProvinceLanzhou 730000, China
| | - Kai Zhang
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Key Laboratory of Bone and Joint Disease Research of Gansu ProvinceLanzhou 730000, China
| | - Ke-Ping Wang
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Lanzhou Xigu District People’s HospitalLanzhou 730000, Gansu, China
| | - Hai-Yu Zhou
- Department of Orthopedics, Lanzhou University Second HospitalLanzhou 730000, Gansu, China
- Lanzhou Xigu District People’s HospitalLanzhou 730000, Gansu, China
| |
Collapse
|
24
|
Lei Y, Junxin C, Yongcan H, Xiaoguang L, Binsheng Y. Role of microRNAs in the crosstalk between osteosarcoma cells and the tumour microenvironment. J Bone Oncol 2020; 25:100322. [PMID: 33083216 PMCID: PMC7554654 DOI: 10.1016/j.jbo.2020.100322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumour, with a peak incidence in adolescents, and the five-year survival rate of patients with metastasis or recurrence is much lower than that of patients without metastasis and recurrence. OS is initiated and develops in a complex tumour microenvironment (TME) that contains many different components, such as osteoblasts, osteoclasts, mesenchymal stem cells, fibroblasts, immune cells, extracellular matrix (ECM), extracellular vesicles, and cytokines. The extensive interaction between OS and the TME underlies OS progression. Therefore, rather than targeting OS cells, targeting the key factors in the TME may yield novel therapeutic approaches. MicroRNAs (miRNAs) play multiple roles in the biological behaviours of OS, and recent studies have implied that miRNAs are involved in mediating the communication between OS cells and the surrounding TME. Here, we review the TME landscape and the miRNA dysregulation of OS, describe the role of the altered TME in OS development and highlight the role of miRNA in the crosstalk between OS cells and the TME.
Collapse
Affiliation(s)
- Yong Lei
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chen Junxin
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Huang Yongcan
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Liu Xiaoguang
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China
| | - Yu Binsheng
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
25
|
uPAR-expressing melanoma exosomes promote angiogenesis by VE-Cadherin, EGFR and uPAR overexpression and rise of ERK1,2 signaling in endothelial cells. Cell Mol Life Sci 2020; 78:3057-3072. [PMID: 33237352 PMCID: PMC8004497 DOI: 10.1007/s00018-020-03707-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/19/2022]
Abstract
Exosomes (Exos) have been reported to promote pre-metastatic niche formation, proliferation, angiogenesis and metastasis. We have investigated the role of uPAR in melanoma cell lines-derived Exos and their pro-angiogenic effects on human microvascular endothelial cells (HMVECs) and endothelial colony-forming cells (ECFCs). Melanoma Exos were isolated from conditioned media of A375 and M6 cells by differential centrifugation and filtration. Tunable Resistive Pulse Sensing (TRPS) and Nanoparticle tracking analysis were performed to analyze dimension and concentration of Exos. The CRISPR–Cas 9 technology was exploited to obtain a robust uPAR knockout. uPAR is expressed in melanoma Exos that are internalized by HMVECs and ECFCs, enhancing VE-Cadherin, EGFR and uPAR expression in endothelial cells that undergo a complete angiogenic program, including proliferation, migration and tube formation. uPAR loss reduced the pro-angiogenic effects of melanoma Exos in vitro and in vivo by inhibition of VE-Cadherin, EGFR and uPAR expression and of ERK1,2 signaling in endothelial cells. A similar effect was obtained with a peptide that inhibits uPAR–EGFR interaction and with the EGFR inhibitor Gefitinib, which also inhibited melanoma Exos-dependent EGFR phosphorylation. This study suggests that uPAR is required for the pro-angiogenic activity of melanoma Exos. We propose the identification of uPAR-expressing Exos as a potentially useful biomarker for assessing pro-angiogenic propensity and eventually monitoring the response to treatment in metastatic melanoma patients.
Collapse
|
26
|
Identification of Kinesin Family Member 2A (KIF2A) as a Promising Therapeutic Target for Osteosarcoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7102757. [PMID: 33204709 PMCID: PMC7655250 DOI: 10.1155/2020/7102757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/07/2019] [Accepted: 08/12/2020] [Indexed: 12/30/2022]
Abstract
Background Osteosarcoma is known as a type of common human bone malignancy, and more therapeutic targets are still required to combat this disease. In recent years, the involvement of KIF2A in cancer progression has been widely revealed; however, its potential effect on osteosarcoma development remains unknown. This study is to assess the KIF2A expression levels in human osteosarcoma tissues and explore its potential role in osteosarcoma development. Methods Immunohistochemical (IHC) assays were conducted to evaluate the expression levels of KIF2A in a total of 74 samples of osteosarcoma tissues and adjacent nontumor tissues. According to the staining intensity in tumor tissues, patients were divided into highly expressed and low expression KIF2A groups. The possible links between the KIF2A expression and the clinical pathological features were explored and analyzed, and the effects of KIF2A on osteosarcoma cell proliferation, migration, and invasion were detected through colony formation assay, MTT assay, wound closure assay, and transwell assay, respectively. The effects of KIF2A on tumor growth and metastasis were detected by the use of animal models. Results KIF2A was highly expressed in human osteosarcoma tissues. Meanwhile, KIF2A was obviously correlated to the tumor size (P = 0.001∗) and clinical stage (P = 0.014∗) of osteosarcoma patients. Our results also revealed that the ablation of KIF2A dramatically blocked the proliferation, migration, and invasion capacity of osteosarcoma cells in vitro and blocked tumor growth and metastasis in mice. Conclusions We investigated the involvement of KIF2A in the development and metastasis of osteosarcoma and therefore thought KIF2A as a promising therapeutic target for osteosarcoma treatment.
Collapse
|
27
|
Wang J, Zhang H, Sun X, Wang X, Ren T, Huang Y, Zhang R, Zheng B, Guo W. Exosomal PD-L1 and N-cadherin predict pulmonary metastasis progression for osteosarcoma patients. J Nanobiotechnology 2020; 18:151. [PMID: 33092576 PMCID: PMC7579953 DOI: 10.1186/s12951-020-00710-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/10/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Recent studies indicated that exosomal programmed death-ligand 1 (PD-L1) derived from cancers could induce immunosuppression and tumor pathogenesis. However, it is unclear how exosomes influence osteosarcoma (OS) progression and whether PD-L1 also exists in serum exosomes (Sr-exosomes) of patients with osteosarcoma. We examined serum exosomes from 70 OS patients, 9 patients with benign tumors and 22 healthy donors. OS-derived exosomes were functionally evaluated in vivo and in vitro. RESULTS The characteristics of exosomes derived from OS patient serum and OS cell lines were confirmed by several methods. We found OS patients had a higher level of exosomal PD-L1 compared to healthy donors. Meanwhile, OS patients with pulmonary metastasis also showed a relatively higher level of exosomal PD-L1 than patients without metastasis. Next, bioinformatic analysis demonstrated that Sr-exosomes isolated from OS patients may involve in the important process of immune function and cancer pathogenesis for OS patients. Co-expression network centered with PD-L1 among Sr-exosomal differently expressed mRNA demonstrated exosomal N-cadherin had a close relationship with exosomal PD-L1 expression. Then, we confirmed higher level of Sr-exosomal N-cadherin in OS patients with pulmonary metastasis compared to ones without metastasis. Furthermore, we elucidated osteosarcoma-derived exosomes and exosomal-PD-L1 promoted the pulmonary metastasis in metastatic models. ROC (Receiver Operating Characteristic Curve) analysis showed AUC (Area Under Curve) of 0.823 for exosomal PD-L1, 0.806 for exosomal N-cadherin and 0.817 for exosomal N-cadherin/E-cadherin to distinguish OS patients with pulmonary metastasis from ones without metastasis. CONCLUSIONS Osteosarcoma stimulates pulmonary metastasis by releasing exosomes, that carry PD-L1 and N-cadherin. Detection of exosomal PD-L1 and N-cadherin from serum of OS patients may predict pulmonary metastasis progression for OS patients.
Collapse
Affiliation(s)
- Jun Wang
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Hongliang Zhang
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Xin Sun
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiaofang Wang
- Beijing Key Laboratory of Bio-Characteristic Profiling for Evaluation of Rational Drug Use, International Cooperation & Joint Laboratoryof Bio-Characteristic Profiling for Evaluation of Rational Drug Use, Beijing Shijitan Hospital, Capital Medical University,, Beijing, 100038, China
| | - Tingting Ren
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Yi Huang
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Ranxin Zhang
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Bingxin Zheng
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Wei Guo
- Peking University People's Hospital, Musculoskeletal Tumor Center, No. 11 Xizhimen South Street, Beijing, 100044, China.
| |
Collapse
|
28
|
Corre I, Verrecchia F, Crenn V, Redini F, Trichet V. The Osteosarcoma Microenvironment: A Complex But Targetable Ecosystem. Cells 2020; 9:cells9040976. [PMID: 32326444 PMCID: PMC7226971 DOI: 10.3390/cells9040976] [Citation(s) in RCA: 257] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/06/2020] [Accepted: 04/11/2020] [Indexed: 01/08/2023] Open
Abstract
Osteosarcomas are the most frequent primary bone sarcomas, affecting mainly children, adolescents, and young adults, and with a second peak of incidence in elderly individuals. The current therapeutic management, a combined regimen of poly-chemotherapy and surgery, still remains largely insufficient, as patient survival has not improved in recent decades. Osteosarcomas are very heterogeneous tumors, both at the intra- and inter-tumor level, with no identified driver mutation. Consequently, efforts to improve treatments using targeted therapies have faced this lack of specific osteosarcoma targets. Nevertheless, these tumors are inextricably linked to their local microenvironment, composed of bone, stromal, vascular and immune cells and the osteosarcoma microenvironment is now considered to be essential and supportive for growth and dissemination. This review describes the different actors of the osteosarcoma microenvironment and gives an overview of the past, current, and future strategies of therapy targeting this complex ecosystem, with a focus on the role of extracellular vesicles and on the emergence of multi-kinase inhibitors.
Collapse
Affiliation(s)
- Isabelle Corre
- INSERM, Nantes University, UMR1238 Phy-Os “Bone Sarcomas and Remodeling of Calcified Tissues”, F-44035 Nantes, France
- CNRS GDR3697 MicroNit, F-37044 Tours, France
- Correspondence: (I.C.); (V.T.)
| | - Franck Verrecchia
- INSERM, Nantes University, UMR1238 Phy-Os “Bone Sarcomas and Remodeling of Calcified Tissues”, F-44035 Nantes, France
| | - Vincent Crenn
- INSERM, Nantes University, UMR1238 Phy-Os “Bone Sarcomas and Remodeling of Calcified Tissues”, F-44035 Nantes, France
- Department of Orthopedic, Nantes Hospital, CHU Hotel-Dieu, F-44035 Nantes, France
| | - Francoise Redini
- INSERM, Nantes University, UMR1238 Phy-Os “Bone Sarcomas and Remodeling of Calcified Tissues”, F-44035 Nantes, France
| | - Valérie Trichet
- INSERM, Nantes University, UMR1238 Phy-Os “Bone Sarcomas and Remodeling of Calcified Tissues”, F-44035 Nantes, France
- CNRS GDR3697 MicroNit, F-37044 Tours, France
- Correspondence: (I.C.); (V.T.)
| |
Collapse
|
29
|
Surman M, Drożdż A, Stępień E, Przybyło M. Extracellular Vesicles as Drug Delivery Systems - Methods of Production and Potential Therapeutic Applications. Curr Pharm Des 2020; 25:132-154. [PMID: 30848183 DOI: 10.2174/1381612825666190306153318] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/01/2019] [Indexed: 01/08/2023]
Abstract
Drug delivery systems are created to achieve the desired therapeutic effect of a specific pharmaceutical compound. Numerous drawbacks and side effects such as unfavorable pharmacokinetics, lack of tissue selectivity, immunogenicity, increased systemic clearance and toxicity, have been observed for currently available drug delivery systems (DDSs). The use of natural and artificial extracellular vesicles (EVs) in drug delivery may help to solve the aforementioned problems faced by different DDSs. Due to their self-origin, small size, flexibility, the presence of multiple adhesive molecules on their surfaces as well as their function as biomolecules carriers, EVs are the perfect candidates for DDSs. Currently, several drug delivery systems based on EVs have been proposed. While the great potential of these particles in targeted drug delivery has been recognized in cancer, hepatitis C, neurodegenerative diseases, inflammatory states etc., this field is still in the early stage of development. Unfortunately, the use of EVs from natural sources (cell cultures, body fluids) results in numerous problems in terms of the heterogeneity of isolated vesicle population as well as the method of isolation thereof, which may influence vesicle composition and properties. Therefore, there is a significant need for the synthesis of artificial EV-based DDSs under strictly controlled laboratory conditions and from well-defined biomolecules (proteins and lipids). Vesicle-mimetic delivery systems, characterized by properties similar to natural EVs, will bring new opportunities to study the mechanisms of DDS internalization and their biological activity after delivering their cargo to a target cell.
Collapse
Affiliation(s)
- Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Anna Drożdż
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Ewa Stępień
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
30
|
Madda R, Chen CM, Wang JY, Chen CF, Chao KY, Yang YM, Wu HY, Chen WM, Wu PK. Proteomic profiling and identification of significant markers from high-grade osteosarcoma after cryotherapy and irradiation. Sci Rep 2020; 10:2105. [PMID: 32034162 PMCID: PMC7005698 DOI: 10.1038/s41598-019-56024-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
Biological reconstruction of allografts and recycled autografts have been widely implemented in high-grade osteogenic sarcoma. For treating tumor-bearing autografts, extracorporeal irradiation (ECIR) and liquid nitrogen (LN) freezing techniques are being used worldwide as a gold standard treatment procedure. Both the methods aim to eradicate the tumor cells from the local recurrence and restore the limb function. Therefore, it is essential and crucial to find, and compare the alterations at molecular and physiological levels of the treated and untreated OGS recycled autografts to obtain valuable clinical information for better clinical practice. Thus, we aimed to investigate the significantly expressed altered proteins from ECIR-and cryotherapy/freezing- treated OGS (n = 12) were compared to untreated OGS (n = 12) samples using LC-ESI-MS/MS analysis, and the selected proteins from this protein panel were verified using immunoblot analysis. From our comparative proteomic analysis identified a total of 131 differentially expressed proteins (DEPs) from OGS. Among these, 91 proteins were up-regulated (2.5 to 3.5-folds), and 40 proteins were down-regulated (0.2 to 0.5 folds) (p < 0.01 and 0.05). The functional enrichment analysis revealed that the identified DEPs have belonged to more than 10 different protein categories include cytoskeletal, extracellular matrix, immune, enzyme modulators, and cell signaling molecules. Among these, we have confirmed two potential candidates’ expressions levels such as Fibronectin and Protein S100 A4 using western blot analysis. Our proteomic study revealed that LN-freezing and ECIR treatments are effectively eradicating tumor cells, and reducing the higher expressions of DEPs at molecular levels which may help in restoring the limb functions of OGS autografts effectively. To the best of our knowledge, this is the first proteomic study that compared proteomic profiles among freezing, ECIR treated with untreated OGS in recycled autografts. Moreover, the verified proteins could be used as prognostic or diagnostic markers that reveal valuable scientific information which may open various therapeutic avenues in clinical practice to improve patient outcomes.
Collapse
Affiliation(s)
- Rashmi Madda
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Research and Development, National Yang-Ming University, Taipei, Taiwan
| | - Chao-Ming Chen
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jir-You Wang
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Fong Chen
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Kuang-Yu Chao
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Min Yang
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Yi Wu
- Instrumentation center, National Taiwan University, Taipei, Taiwan
| | - Wei-Ming Chen
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Kuei Wu
- Department of Orthopedics & Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan. .,Department of orthopedics, Therapeutical and Musculoskeletal Tumor Research Center, Taipei Veterans General Hospital, Taipei, Taiwan. .,Orthopedic Department, School of Medicine, National Yang-Ming University, Taipei, Taiwan. .,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
31
|
Chicón-Bosch M, Tirado OM. Exosomes in Bone Sarcomas: Key Players in Metastasis. Cells 2020; 9:cells9010241. [PMID: 31963599 PMCID: PMC7016778 DOI: 10.3390/cells9010241] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/11/2020] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Bone sarcomas are rare cancers which often present with metastatic disease and are still associated with poor survival rates. Studies in the last decade have identified that exosomes, a type of extracellular vesicle released by cells, play an important role in tumour progression and dissemination. Through the transfer of their cargo (RNAs, proteins, and lipids) across cells, they are involved in cellular cross-talk and can induce changes in cellular behaviour. Exosomes have been shown to be important in metastasis organotropism, induction of angiogenesis and vascular permeability, the education of cells towards a pro-metastatic phenotype or the interaction between stromal and tumour cells. Due to the importance exosomes have in disease progression and the high incidence of metastasis in bone sarcomas, recent studies have evaluated the implications of these extracellular vesicles in bone sarcomas. In this review, we discuss the studies that evaluate the role of exosomes in osteosarcoma, Ewing sarcoma, and preliminary data on chondrosarcoma.
Collapse
Affiliation(s)
- Mariona Chicón-Bosch
- Sarcoma Research Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- Correspondence: (M.C.-B.); (O.M.T.); Tel.: +34-9326-0742 (M.C.-B.); +34-932-603-823 (O.M.T.)
| | - Oscar M. Tirado
- Sarcoma Research Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- CIBERONC, Carlos III Institute of Health (ISCIII), 28029 Madrid, Spain
- Institut Català d’Oncologia (ICO), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- Correspondence: (M.C.-B.); (O.M.T.); Tel.: +34-9326-0742 (M.C.-B.); +34-932-603-823 (O.M.T.)
| |
Collapse
|
32
|
Nanomedicine in osteosarcoma therapy: Micelleplexes for delivery of nucleic acids and drugs toward osteosarcoma-targeted therapies. Eur J Pharm Biopharm 2020; 148:88-106. [PMID: 31958514 DOI: 10.1016/j.ejpb.2019.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/09/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023]
Abstract
Osteosarcoma(OS) represents the main cancer affecting bone tissue, and one of the most frequent in children. In this review we discuss the major pathological hallmarks of this pathology, its current therapeutics, new active biomolecules, as well as the nanotechnology outbreak applied to the development of innovative strategies for selective OS targeting. Small RNA molecules play a role as key-regulator molecules capable of orchestrate different responses in what concerns cancer initiation, proliferation, migration and invasiveness. Frequently associated with lung metastasis, new strategies are urgent to upgrade the therapeutic outcomes and the life-expectancy prospects. Hence, the prominent rise of micelleplexes as multifaceted and efficient structures for nucleic acid delivery and selective drug targeting is revisited here with special emphasis on ligand-mediated active targeting. Future landmarks toward the development of novel nanostrategies for both OS diagnosis and OS therapy improvements are also discussed.
Collapse
|
33
|
Kok VC, Yu CC. Cancer-Derived Exosomes: Their Role in Cancer Biology and Biomarker Development. Int J Nanomedicine 2020; 15:8019-8036. [PMID: 33116515 PMCID: PMC7585279 DOI: 10.2147/ijn.s272378] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/29/2020] [Indexed: 02/05/2023] Open
Abstract
Exosomes are a subset of tiny extracellular vesicles manufactured by all cells and are present in all body fluids. They are produced actively in tumor cells, which are released and utilized to facilitate tumor growth. Their characteristics enable them to assist major cancer hallmarks, leveraged by cancer cells in fostering cancer growth and spread while implementing ways to escape elimination from the host environment. This review updates on the latest progress on the roles of cancer-derived exosomes, of 30-100 nm in size, in deregulating paracrine trafficking in the tumor microenvironment and circulation. Thus, exosomes are being exploited in diagnostic biomarker development, with its potential in clinical applications as therapeutic targets utilized in exosome-based nanoparticle drug delivery strategies for cancer therapy. Ongoing studies were retrieved from PubMed® and Scopus database and ClinicalTrials.gov registry for review, highlighting how cancer cells from entirely different cell lines rely on genetic information carried by their exosomes for homotypic and heterotypic intercellular communications in the microenvironment to favor proliferation and invasion, while establishing a pre-metastatic niche in welcoming cancer cells' arrival. We will elaborate on the trafficking of tumor-derived exosomes in fostering cancer proliferation, invasion, and metastasis in hematopoietic (leukemia and myeloma), epithelial (breast cancer), and mesenchymal (soft tissue sarcoma and osteosarcoma) cancers. Cancer-derived exosomal trafficking is observed in several types of liquid or solid tumors, confirming their role as cancer hallmark enabler. Their enriched genetic signals arising from their characteristic DNA, RNA, microRNA, and lncRNA, along with specific gene expression profiles, protein, or lipid composition carried by the exosomal cargo shed into blood, saliva, urine, ascites, and cervicovaginal lavage, are being studied as a diagnostic, prognostic, or predictive cancer biomarker. We reveal the latest research efforts in exploiting the use of nanoparticles to improve the overall cancer diagnostic capability in the clinic.
Collapse
Affiliation(s)
- Victor C Kok
- Division of Medical Oncology, Kuang Tien General Hospital Cancer Center, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Correspondence: Victor C Kok Division of Medical Oncology, Kuang Tien General Hospital Cancer Center, 117 Shatien Road, Shalu, Taichung43303, TaiwanTel +886 4 2662 5111 Ext 2263Fax +886 4 2665 5050 Email
| | - Cheng-Chia Yu
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
34
|
Exosomes: Dynamic Mediators of Extracellular Communication in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:189-197. [PMID: 32767243 DOI: 10.1007/978-3-030-43085-6_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is becoming increasingly recognized that the tumor microenvironment significantly contributes to the development, progression, and metastasis of cancer and also plays a role in response to treatment. The tumor microenvironment is a complex and heterogeneous niche comprised of stromal cells, cancer cells, blood vessels, areas of hypoxia and necrotic tissue, fibrosis, and extracellular matrix. Cellular communication takes place within the tumor microenvironment, both via cell to cell contact, and through extracellular mechanisms such as exosomal signaling. Exosomes are very small membrane-bound vesicles that have been shown to play key roles in the progression of cancer including modulation of the tumor microenvironment through the induction of angiogenesis, the transfer of genetic information that confers drug resistance, and increased cell migration, invasion, proliferation, and survival, as well as the modulation of immune cell interactions. The role of exosomes in several different cancers has been investigated. In the context of osteosarcoma, understanding how exosomes may modulate the tumor microenvironment to support metastatic growth particularly in the lung, the most common site of metastases, may identify novel therapeutic targets for relapsed patients.
Collapse
|
35
|
Perut F, Roncuzzi L, Baldini N. The Emerging Roles of Extracellular Vesicles in Osteosarcoma. Front Oncol 2019; 9:1342. [PMID: 31850225 PMCID: PMC6901498 DOI: 10.3389/fonc.2019.01342] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous nanosized vesicles that are constitutively released by virtually all types of cells. They have been isolated in almost all body fluids. EVs cargo consists of various molecules (nucleic acids, proteins, lipids, and metabolites), that can be found on EVs surface and/or in their lumen. EVs structure confer stability and allow the transfer of their cargo to specific cell types over a distance. EVs play a critical role in intercellular communication in physiological and pathological settings. The broadening of knowledge on EVs improved our comprehension of cancer biology as far as tumor development, growth, metastasis, chemoresistance, and treatment are concerned. Increasing evidences suggest that EVs have a significant role in osteosarcoma (OS) development, progression, and metastatic process. The modulation of inflammatory communication pathways by EVs plays a critical role in OS and in other bone-related pathological conditions such as osteoarthritis and rheumatoid arthritis. In this review we describe the emerging data on the role of extracellular vesicles in osteosarcoma and discuss the effects and function of OS-derived EVs focusing on their future applicability in clinical practice.
Collapse
Affiliation(s)
- Francesca Perut
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Laura Roncuzzi
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
36
|
Endo-Munoz L, Bennett TC, Topkas E, Wu SY, Thamm DH, Brockley L, Cooper M, Sommerville S, Thomson M, O'Connell K, Lane A, Bird G, Peaston A, Matigian N, Straw RC, Saunders NA. Auranofin improves overall survival when combined with standard of care in a pilot study involving dogs with osteosarcoma. Vet Comp Oncol 2019; 18:206-213. [PMID: 31441983 DOI: 10.1111/vco.12533] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/11/2019] [Accepted: 08/16/2019] [Indexed: 12/29/2022]
Abstract
Osteosarcoma is the most common paediatric primary bone malignancy. The major cause of death in osteosarcoma is drug-resistant pulmonary metastasis. Previous studies have shown that thioredoxin reductase 2 is a driver of metastasis in osteosarcoma and can be inhibited by auranofin (AF). Moreover, studies have shown that AF significantly reduces pulmonary metastases in xenotransplant models. Here, we describe a phase I/II study of AF in canine osteosarcoma, a well-recognized spontaneous model of human osteosarcoma. We performed a single-arm multicentre pilot study of AF in combination with standard of care (SOC) (amputation + carboplatin). We recruited 40 dogs to the trial and used a historical SOC-only control group (n = 26). Dogs >15 kg received 9 mg AF q3d PO and dogs <15 kg received 6 mg q3d. Follow-up occurred over at least a 3-year period. Auranofin plus SOC improved overall survival (OS) (P = .036) in all dogs treated. The improved outcome was attributable entirely to improved OS in male dogs (P = .009). At the time of writing, 10 dogs (25%) survive without measurable disease in the treatment group with survival times ranging between 806 and 1525 days. Our study shows that AF improves OS in male dogs when combined with SOC. Our findings have translational relevance for the management of canine and human osteosarcoma. Our data justify a larger multicentre phase 2 trial in dogs and a phase I/II trial in human patients with refractory disease at the time of initial surgery.
Collapse
Affiliation(s)
- Liliana Endo-Munoz
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia.,Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | | | - Eleni Topkas
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Sherry Y Wu
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Douglas H Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Laura Brockley
- Victorian Animal Cancer Care, Melbourne, Victoria, Australia
| | - Maureen Cooper
- Victorian Animal Cancer Care, Melbourne, Victoria, Australia
| | - Scott Sommerville
- Department of Orthopedic Oncology, Princess Alexandra Hospital and The Wesley Hospital, Brisbane, Queensland, Australia
| | - Maurine Thomson
- Veterinary Specialist Services, Brisbane, Queensland, Australia
| | | | - Amy Lane
- Small Animal Oncology, Newcastle, New South Wales, Australia
| | - Guy Bird
- Veterinary Emergency Centre and Hospital, James Cook University School of Veterinary and Biomedical Science, Townsville, Queensland, Australia
| | - Anne Peaston
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nicholas Matigian
- QFAB Bioinformatics, BIODATA Institute of Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Rodney C Straw
- Brisbane Veterinary Specialist Centre, Brisbane, Queensland, Australia.,Australian Consortium of Comparative Oncology of the Australian Animal Cancer Foundation, Brisbane, Queensland, Australia
| | - Nicholas A Saunders
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
37
|
Cappariello A, Rucci N. Tumour-Derived Extracellular Vesicles (EVs): A Dangerous "Message in A Bottle" for Bone. Int J Mol Sci 2019; 20:E4805. [PMID: 31569680 PMCID: PMC6802008 DOI: 10.3390/ijms20194805] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
Several studies have shown the importance of Extracellular Vesicles (EVs) in the intercellular communication between tumour and resident cells. Through EVs, tumour cells can trigger cell-signalling molecules and shuttle exogenous information to target cells, thus promoting spread of the disease. In fact, many processes are fuelled by EVs, such as tumour invasion and dormancy, drug-resistance, immune-surveillance escape, extravasation, extracellular matrix remodelling and metastasis. A key element is certainly the molecular profile of the shed cargo. Understanding the biochemical basis of EVs would help to predict the ability and propensity of cancer cells to metastasize a specific tissue, with the aim to target the release of EVs and to manipulate their content as a possible therapeutic approach. Moreover, EV profiling could help monitor the progression of cancer, providing a useful tool for more effective therapy. This review will focus on all the EV-mediated mentioned mechanisms in the context of both primary bone cancers and bone metastases.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Department of Onco-haematology IRCCS Bambino Gesù Children's Hospital, 00152 Rome, Italy.
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| |
Collapse
|
38
|
Jin Y, Long D, Li J, Yu R, Song Y, Fang J, Yang X, Zhou S, Huang S, Zhao Z. Extracellular vesicles in bone and tooth: A state-of-art paradigm in skeletal regeneration. J Cell Physiol 2019; 234:14838-14851. [PMID: 30847902 DOI: 10.1002/jcp.28303] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 02/05/2023]
Abstract
Bone and tooth, fundamental parts of the craniofacial skeleton, are anatomically and developmentally interconnected structures. Notably, pathological processes in these tissues underwent together and progressed in multilevels. Extracellular vesicles (EVs) are cell-released small organelles and transfer proteins and genetic information into cells and tissues. Although EVs have been identified in bone and tooth, particularly EVs have been identified in the bone formation and resorption, the concrete roles of EVs in bone and tooth development and diseases remain elusive. As such, we review the recent progress of EVs in bone and tooth to highlight the novel findings of EVs in cellular communication, tissue homeostasis, and interventions. This will enhance our comprehension on the skeletal biology and shed new light on the modulation of skeletal disorders and the potential of genetic treatment.
Collapse
Affiliation(s)
- Ying Jin
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Juan Li
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Ruichao Yu
- Department of Pulmonary, Brigham and Women's Hospital, Harvard Medical School, Massachusetts
| | - Yueming Song
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Fang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Xi Yang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shu Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, P.R. China
| | - Shishu Huang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
39
|
Protein Kinase B and Extracellular Signal-Regulated Kinase Inactivation is Associated with Regorafenib-Induced Inhibition of Osteosarcoma Progression In Vitro and In Vivo. J Clin Med 2019; 8:jcm8060900. [PMID: 31238539 PMCID: PMC6616516 DOI: 10.3390/jcm8060900] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma is the most common type of bone cancer. Multimodality treatment involving chemotherapy, radiotherapy and surgery is not effective enough to control osteosarcoma. Regorafenib, the oral multi-kinase inhibitor, has been shown to have positive efficacy on disease progression delay in chemotherapy resistant osteosarcoma patients. However anti-cancer effect and mechanism of regorafenib in osteosarcoma is ambiguous. Thus, the aim of this study is to investigate the efficacy and molecular mechanism of regorafenib on osteosarcoma in vitro and in vivo. Human osteosarcomas U-2 OS or MG-63 were treated with regorafenib, miltefosine (protein kinase B (AKT) inhibitor), or PD98059 (mitogen-activated protein/extracellular signal-regulated kinase (MEK) pathway inhibitor) for 24 or 48 h. Cell viability, apoptotic signaling transduction, tumor invasion, expression of tumor progression-associated proteins and tumor growth after regorafenib treatment were assayed by MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, flow cytometry, transwell assay, Western blotting assay and in vivo animal experiment, respectively. In these studies, we also indicated that regorafenib suppressed cell growth by prompting apoptosis of osteosarcoma cells, which is mediated through inactivation of ERK and AKT signaling pathways. After regorafenib treatment, downregulation of related genes in invasion (vascular endothelial growth factor (VEGF) and matrix metallopeptidase 9 (MMP-9)), proliferation (CyclinD1) and anti-apoptosis (X-linked inhibitor of apoptosis protein (XIAP), myeloid cell leukemia-1 (MCL-1), and cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein (C-FLIP)) were found. Moreover, upregulation of caspase-3 and caspase-8 cleavage were also observed. In sum, we suggest that regorafenib has potential to suppress osteosarcoma progression via inactivation of AKT and ERK mediated signaling pathway.
Collapse
|
40
|
Perut F, Roncuzzi L, Zini N, Massa A, Baldini N. Extracellular Nanovesicles Secreted by Human Osteosarcoma Cells Promote Angiogenesis. Cancers (Basel) 2019; 11:cancers11060779. [PMID: 31195680 PMCID: PMC6627280 DOI: 10.3390/cancers11060779] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/28/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis involves a number of different players among which extracellular nanovesicles (EVs) have recently been proposed as an efficient cargo of pro-angiogenic mediators. Angiogenesis plays a key role in osteosarcoma (OS) development and progression. Acidity is a hallmark of malignancy in a variety of cancers, including sarcomas, as a result of an increased energetic metabolism. The aim of this study was to investigate the role of EVs derived from osteosarcoma cells on angiogenesis and whether extracellular acidity, generated by tumor metabolism, could influence EVs activity. For this purpose, we purified and characterized EVs from OS cells maintained at either acidic or neutral pH. The ability of EVs to induce angiogenesis was assessed in vitro by endothelial cell tube formation and in vivo using chicken chorioallantoic membrane. Our findings demonstrated that EVs derived from osteosarcoma cells maintained either in acidic or neutral conditions induced angiogenesis. The results showed that miRNA and protein content of EVs cargo are correlated with pro-angiogenic activity and this activity is increased by the acidity of tumor microenvironment. This study provides evidence that EVs released by human osteosarcoma cells act as carriers of active angiogenic stimuli that are able to promote endothelial cell functions relevant to angiogenesis.
Collapse
Affiliation(s)
- Francesca Perut
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Laura Roncuzzi
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Nicoletta Zini
- CNR-National Research Council of Italy, Institute of Molecular Genetics, 40136 Bologna, Italy.
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Annamaria Massa
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Nicola Baldini
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40123 Bologna, Italy.
| |
Collapse
|
41
|
Masaoutis C, Korkolopoulou P, Theocharis S. Exosomes in sarcomas: Tiny messengers with broad implications in diagnosis, surveillance, prognosis and treatment. Cancer Lett 2019; 449:172-177. [PMID: 30779943 DOI: 10.1016/j.canlet.2019.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 02/08/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022]
Abstract
Exosomes are cell-secreted extracellular vesicles, which contain an array of biomolecules, such as proteins, mRNAs, microRNAs, and lipids, take part in intercellular communication and mediate tumor-host interactions. They are increasingly considered as a source of biomarkers for liquid biopsies as well as potential drug vectors. Sarcomas are rare malignant mesenchymal tumours and due to their relative rarity exosomes have not been investigated in as extensively as in epithelial malignancies. Nonetheless, valuable information has been gathered over the last years on the roles of exosomes in sarcomas. In the present review we summarize all relevant data obtained so far from cell lines, animal models and patients with emphasis on their potential clinical utility.
Collapse
Affiliation(s)
- Christos Masaoutis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Penelope Korkolopoulou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| |
Collapse
|
42
|
Poon AC, Inkol JM, Luu AK, Mutsaers AJ. Effects of the potassium-sparing diuretic amiloride on chemotherapy response in canine osteosarcoma cells. J Vet Intern Med 2018; 33:800-811. [PMID: 30556178 PMCID: PMC6430882 DOI: 10.1111/jvim.15382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023] Open
Abstract
Background Osteosarcoma (OSA) is a common bone tumor of mesenchymal origin in dogs. Chemotherapy delays metastasis, yet most dogs die of this disease within 1 year of diagnosis. The high metabolic demand of cancer cells promotes proton pump upregulation, leading to acidification of the tumor microenvironment and chemoresistance. The potassium‐sparing diuretic amiloride is among a class of proton pump inhibitors prescribed for refractory heart failure treatment in dogs. Objective We hypothesized that amiloride treatment improves chemotherapy response by reducing acidification in canine OSA cells. Our objective was to assess the in vitro effects of amiloride on cell viability, apoptosis, and metabolism. Methods In vitro study. Assessments of cell viability and apoptosis were performed after single agent or combination treatment, along with calculations of pharmacological synergism using the combination index. Protein signaling during apoptosis was evaluated by Western blotting. Metabolic profiling was performed using a Seahorse bioanalyzer. Results Amiloride strongly synergized with doxorubicin in combination treatment and exhibited additive or antagonistic effects with carboplatin in canine OSA cells. Combination treatment with doxorubicin significantly upregulated p53‐mitochondrial signaling to activate apoptosis and downregulate Akt phosphorylation. Amiloride‐treated cells further exhibited metabolic switching with reductions in glycolytic capacity and maximal respiration. Conclusion and Clinical Importance Amiloride synergized with doxorubicin to potentiate apoptosis in canine OSA cells. These results justify further investigation into repurposing of amiloride as an oncology drug for the treatment of OSA in dogs.
Collapse
Affiliation(s)
- Andrew C Poon
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jordon M Inkol
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Anita K Luu
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Anthony J Mutsaers
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.,Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
43
|
Mahmood N, Mihalcioiu C, Rabbani SA. Multifaceted Role of the Urokinase-Type Plasminogen Activator (uPA) and Its Receptor (uPAR): Diagnostic, Prognostic, and Therapeutic Applications. Front Oncol 2018; 8:24. [PMID: 29484286 PMCID: PMC5816037 DOI: 10.3389/fonc.2018.00024] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/24/2018] [Indexed: 01/01/2023] Open
Abstract
The plasminogen activator (PA) system is an extracellular proteolytic enzyme system associated with various physiological and pathophysiological processes. A large body of evidence support that among the various components of the PA system, urokinase-type plasminogen activator (uPA), its receptor (uPAR), and plasminogen activator inhibitor-1 and -2 (PAI-1 and PAI-2) play a major role in tumor progression and metastasis. The binding of uPA with uPAR is instrumental for the activation of plasminogen to plasmin, which in turn initiates a series of proteolytic cascade to degrade the components of the extracellular matrix, and thereby, cause tumor cell migration from the primary site of origin to a distant secondary organ. The components of the PA system show altered expression patterns in several common malignancies, which have identified them as ideal diagnostic, prognostic, and therapeutic targets to reduce cancer-associated morbidity and mortality. This review summarizes the various components of the PA system and focuses on the role of uPA-uPAR in different biological processes especially in the context of malignancy. We also discuss the current state of knowledge of uPA-uPAR-targeted diagnostic and therapeutic strategies for various malignancies.
Collapse
Affiliation(s)
- Niaz Mahmood
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Catalin Mihalcioiu
- Department of Oncology, McGill University Health Centre, Montreal, QC, Canada
| | - Shafaat A. Rabbani
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Department of Oncology, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
44
|
Diao C, Xi Y, Xiao T. Identification and analysis of key genes in osteosarcoma using bioinformatics. Oncol Lett 2017; 15:2789-2794. [PMID: 29435005 PMCID: PMC5778824 DOI: 10.3892/ol.2017.7649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 06/21/2017] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma (OS) is an invasive malignant neoplasm of the bones. The present study identified and analyzed key genes associated with OS. Expression profiling of the dataset GSE49003, which included 6 metastatic and 6 non-metastatic OS cell lines and was obtained from the Gene Expression Omnibus, was performed. Following data preprocessing, the differentially expressed genes (DEGs) were selected using the limma package in R. Subsequently, bidirectional hierarchical clustering using the pheatmap package in R and an unpaired Students' t-test was performed for the DEGs. Based on the Search Tool for the Retrieval of Interacting Genes database and Cytoscape software, a protein-protein interaction (PPI) network for the DEGs was constructed. Using Database for Annotation, Visualization and Integrated Discovery software and the Kyoto Encyclopedia of Genes and Genomes Orthology Based Annotation System server, functional and pathway enrichment analyses were performed for the DEGs corresponding to the proteins of the network. In addition, the transcription factors (TFs) and CpG islands of the gene promoter were searched for using the TRANSFAC database and CpG Island Searcher software, respectively. A total of 323 DEGs were identified between the metastatic and non-metastatic samples. In the PPI network, upregulated epidermal growth factor receptor (EGFR) exhibits a high degree and was therefore highly interconnected with other proteins. Enrichment analysis revealed that EGFR was enriched in cytoskeleton organization, organic substance response and the signaling pathway of focal adhesion. The TFs early growth response 1, nuclear factor-κB complex subunits, peroxisome proliferator activated receptor α, signal transducer and activator of transcription 3 and MYC proto-oncogene were identified in the EGFR promoter region. Furthermore, multiple CpG islands, starting from the 400 bp of the EGFR promoter sequence, were predicted. Methylated modification of the CpG islands in the EGFR promoter may help to regulate EGFR expression. The TFs identified in the EGFR promoter may function in the progression of OS.
Collapse
Affiliation(s)
- Chunyu Diao
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yong Xi
- Department of Orthopedics, Tongchuan People's Hospital, Tongchuan, Shaanxi 727000, P.R. China
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
45
|
Abstract
Better diagnostic biomarkers and therapeutic options are still necessary for patients with sarcomas due to the current limitations of diagnosis and treatment. Exosomes are small extracellular membrane vesicles that are released by various cells and are found in most body fluids. Tumor-derived exosomes have been proven to mediate tumorigenesis, intercellular communication, microenvironment modulation, and metastasis in different cancers, including in sarcomas. Recently, exosomes have been considered as potential biomarkers for sarcoma diagnosis and prognosis, and as possible targets for sarcoma therapy. Moreover, due to their specific cell tropism and bioavailability, exosomes can also be engineered as vehicles for drug delivery. In this review, we discuss recent advances in the roles of tumor-derived exosomes in sarcoma and their potential clinical applications.
Collapse
|
46
|
Re-calculating! Navigating through the osteosarcoma treatment roadblock. Pharmacol Res 2016; 117:54-64. [PMID: 27940205 DOI: 10.1016/j.phrs.2016.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 01/03/2023]
Abstract
The survival rates for patients with osteosarcoma have remained almost static for the past three decades. Current standard of care therapy includes chemotherapies such as doxorubicin, cisplatin, and methotrexate along with complete surgical resection and surgery with or without ifosfamide and etoposide for relapse, though outcomes are hoped to be improved through clinical trials. Additionally, increased understanding of the genetics, signaling pathways and microenvironmental factors driving the disease have led to the identification of promising agents and potential paths towards translation of an exciting array of novel targeted therapies. Here, we review the mechanism of action of these emerging therapies and how, with clinical translation, they can potentially improve the survival rates for osteosarcoma patients in the near future.
Collapse
|
47
|
Shen J, Meyers CA, Shrestha S, Singh A, LaChaud G, Nguyen V, Asatrian G, Federman N, Bernthal N, Eilber FC, Dry SM, Ting K, Soo C, James AW. Sclerostin expression in skeletal sarcomas. Hum Pathol 2016; 58:24-34. [PMID: 27498059 DOI: 10.1016/j.humpath.2016.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023]
Abstract
Sclerostin (SOST) is an extracellular Wnt signaling antagonist which negatively regulates bone mass. Despite this, the expression and function of SOST in skeletal tumors remain poorly described. Here, we first describe the immunohistochemical staining pattern of SOST across benign and malignant skeletal tumors with bone or cartilage matrix (n=68 primary tumors). Next, relative SOST expression was compared to markers of Wnt signaling activity and osteogenic differentiation across human osteosarcoma (OS) cell lines (n=7 cell lines examined). Results showed immunohistochemical detection of SOST in most bone-forming tumors (90.2%; 46/51) and all cartilage-forming tumors (100%; 17/17). Among OSs, variable intensity and distribution of SOST expression were observed, which highly correlated with the presence and degree of neoplastic bone. Patchy SOST expression was observed in cartilage-forming tumors, which did not distinguish between benign and malignant tumors or correlate with regional morphologic characteristics. Finally, SOST expression varied widely between OS cell lines, with more than 97-fold variation. Among OS cell lines, SOST expression positively correlated with the marker of osteogenic differentiation alkaline phosphatase and did not correlate well with markers of Wnt/β-catenin signaling activity. In summary, SOST is frequently expressed in skeletal bone- and cartilage-forming tumors. The strong spatial correlation with bone formation and the in vitro expression patterns are in line with the known functions of SOST in nonneoplastic bone, as a feedback inhibitor on osteogenic differentiation. With anti-SOST as a potential therapy for osteoporosis in the near future, its basic biologic and phenotypic consequences in skeletal tumors should not be overlooked.
Collapse
Affiliation(s)
- Jia Shen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA 90095; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Carolyn A Meyers
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Swati Shrestha
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Arun Singh
- Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Greg LaChaud
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA 90095; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Vi Nguyen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Greg Asatrian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA 90095; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Noah Federman
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Nicholas Bernthal
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA 90095
| | - Fritz C Eilber
- Division of Surgical Oncology, University of California, Los Angeles, CA 90095
| | - Sarah M Dry
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA 90095
| | - Chia Soo
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA 90095; Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Aaron W James
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095; Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095; Department of Pathology, Johns Hopkins University, Baltimore, MD 21205.
| |
Collapse
|
48
|
Macklin R, Wang H, Loo D, Martin S, Cumming A, Cai N, Lane R, Ponce NS, Topkas E, Inder K, Saunders NA, Endo-Munoz L. Extracellular vesicles secreted by highly metastatic clonal variants of osteosarcoma preferentially localize to the lungs and induce metastatic behaviour in poorly metastatic clones. Oncotarget 2016; 7:43570-43587. [PMID: 27259278 PMCID: PMC5190045 DOI: 10.18632/oncotarget.9781] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/25/2016] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) is the most common pediatric bone tumor and is associated with the emergence of pulmonary metastasis. Unfortunately, the mechanistic basis for metastasis remains unclear. Tumor-derived extracellular vesicles (EVs) have been shown to play critical roles in cell-to-cell communication and metastatic progression in other cancers, but their role in OS has not been explored. We show that EVs secreted by cells derived from a highly metastatic clonal variant of the KHOS cell line can be internalized by a poorly metastatic clonal variant of the same cell line and induce a migratory and invasive phenotype. This horizontal phenotypic transfer is unidirectional and provides evidence that metastatic potential may arise via interclonal co-operation. Proteomic analysis of the EVs secreted by highly metastatic OS clonal variants results in the identification of a number of proteins and G-protein coupled receptor signaling events as potential drivers of OS metastasis and novel therapeutic targets. Finally, multiphoton microscopy with fluorescence lifetime imaging in vivo, demonstrated a preferential seeding of lung tissue by EVs derived from highly metastatic OS clonal variants. Thus, we show that EVs derived from highly metastatic clonal variants of OS may drive metastatic behaviour via interclonal co-operation and preferential colonization of the lungs.
Collapse
Affiliation(s)
- Rebecca Macklin
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Haolu Wang
- Therapeutics Research Centre, School of Medicine, University of Queensland, Brisbane, Australia
| | - Dorothy Loo
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Sally Martin
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Andrew Cumming
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Na Cai
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Rebecca Lane
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Natalia Saenz Ponce
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Eleni Topkas
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Kerry Inder
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Nicholas A Saunders
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Liliana Endo-Munoz
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| |
Collapse
|