1
|
Wen Y, Zhang X, Zhang J, Lu Z. Deciphering the role of lipid metabolism and acetylation in osteosarcoma: A comprehensive molecular analysis. ENVIRONMENTAL TOXICOLOGY 2024; 39:4776-4790. [PMID: 39162397 DOI: 10.1002/tox.24325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 08/21/2024]
Abstract
Osteosarcoma, known for its rapid progression and high metastatic potential, poses significant challenges in adolescent oncology. This study delves into the roles of lipid metabolism and acetylation genes in the disease's pathogenesis. Utilizing gene set variation analysis, we examined 14 lipid metabolism-related pathways in osteosarcoma patients, identifying significant variances in three pathways between metastatic and primary cases. Additionally, differences in four acetylation genes between these groups were observed. A comprehensive analysis pinpointed 62 lipid metabolism-related genes, with 39 exhibiting significant correlations with acetylation genes, termed lipid metabolism acetylation (LMA) genes. Employing machine learning techniques like Lasso+RSF and GBM, we developed a predictive model for overall survival based on LMA genes. This model, with an average c-index of 0.771, focuses on three key genes: CYP2C8, PAFAH2, and ACOX3, whose prognostic value was confirmed through survival and receiver operating characteristic curve analyses. Quantitative RT-PCR results indicated higher expression levels of ACOX3 and PAFAH2 in OS cells (143B, HOS, MG63) than in osteoblasts (hFOB1.19), while CYP2C8 was lower in OS cells. Furthermore, drug sensitivity analysis through the pRRophetic algorithm suggested potential targeted therapies, revealing drugs with differential sensitivity based on LMA scores and varied treatment responses related to the expression of core genes. This study not only highlights the crucial role of lipid metabolism and acetylation in osteosarcoma but also offers a foundation for personalized treatment strategies, marking a notable advancement in combating this severe form of adolescent cancer.
Collapse
Affiliation(s)
- Yong Wen
- Department of Orthopedics, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xijiang Zhang
- Department of Intensive Care Unit, Taizhou Municipal Hospital, Taizhou, China
| | - Jin Zhang
- Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou, China
| | - Zhisheng Lu
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
2
|
Janssen FW, Lak NSM, Janda CY, Kester LA, Meister MT, Merks JHM, van den Heuvel-Eibrink MM, van Noesel MM, Zsiros J, Tytgat GAM, Looijenga LHJ. A comprehensive overview of liquid biopsy applications in pediatric solid tumors. NPJ Precis Oncol 2024; 8:172. [PMID: 39097671 PMCID: PMC11297996 DOI: 10.1038/s41698-024-00657-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/15/2024] [Indexed: 08/05/2024] Open
Abstract
Liquid biopsies are emerging as an alternative source for pediatric cancer biomarkers with potential applications during all stages of patient care, from diagnosis to long-term follow-up. While developments within this field are reported, these mainly focus on dedicated items such as a specific liquid biopsy matrix, analyte, and/or single tumor type. To the best of our knowledge, a comprehensive overview is lacking. Here, we review the current state of liquid biopsy research for the most common non-central nervous system pediatric solid tumors. These include neuroblastoma, renal tumors, germ cell tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma and other soft tissue sarcomas, and liver tumors. Within this selection, we discuss the most important or recent studies involving liquid biopsy-based biomarkers, anticipated clinical applications, and the current challenges for success. Furthermore, we provide an overview of liquid biopsy-based biomarker publication output for each tumor type based on a comprehensive literature search between 1989 and 2023. Per study identified, we list the relevant liquid biopsy-based biomarkers, matrices (e.g., peripheral blood, bone marrow, or cerebrospinal fluid), analytes (e.g., circulating cell-free and tumor DNA, microRNAs, and circulating tumor cells), methods (e.g., digital droplet PCR and next-generation sequencing), the involved pediatric patient cohort, and proposed applications. As such, we identified 344 unique publications. Taken together, while the liquid biopsy field in pediatric oncology is still behind adult oncology, potentially relevant publications have increased over the last decade. Importantly, steps towards clinical implementation are rapidly gaining ground, notably through validation of liquid biopsy-based biomarkers in pediatric clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Michael T Meister
- Princess Máxima Center, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Johannes H M Merks
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center, Utrecht, the Netherlands
- Wilhelmina Children's Hospital-Division of CHILDHEALTH, University Medical Center Utrech, University of Utrecht, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | | | - Godelieve A M Tytgat
- Princess Máxima Center, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Leendert H J Looijenga
- Princess Máxima Center, Utrecht, the Netherlands.
- Department of Pathology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Zhai C, Xu J, Yang Y, Xie F, Cao L, Wang K, Zhou Y, Ding X, Yin J, Ding X, Hu H, Yu H. Heterogeneous Analysis of Extracellular Vesicles for Osteosarcoma Diagnosis. Anal Chem 2024; 96:9486-9492. [PMID: 38814722 DOI: 10.1021/acs.analchem.4c00941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Osteosarcoma (OS) is the most prevalent primary tumor of bones, often diagnosed late with a poor prognosis. Currently, few effective biomarkers or diagnostic methods have been developed for early OS detection with high confidence, especially for metastatic OS. Tumor-derived extracellular vesicles (EVs) are emerging as promising biomarkers for early cancer diagnosis through liquid biopsy. Here, we report a plasmonic imaging-based biosensing technique, termed subpopulation protein analysis by single EV counting (SPASEC), for size-dependent EV subpopulation analysis. In our SPASEC platform, EVs are accurately sized and counted on plasmonic sensor chips coated with OS-specific antibodies. Subsequently, EVs are categorized into distinct subpopulations based on their sizes, and the membrane proteins of each size-dependent subpopulation are profiled. We measured the heterogeneous expression levels of the EV markers (CD63, BMP2, GD2, and N-cadherin) in each of the EV subsets from both OS cell lines and clinical plasma samples. Using the linear discriminant analysis (LDA) model, the combination of four markers is applied to classify the healthy donors (n = 37), nonmetastatic OS patients (n = 13), and metastatic patients (n = 12) with an area under the curve of 0.95, 0.92, and 0.99, respectively. SPASEC provides accurate EV sensing technology for early OS diagnosis.
Collapse
Affiliation(s)
- Chunhui Zhai
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jiaying Xu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yuting Yang
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Feng Xie
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Li Cao
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Kai Wang
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Zhou
- Department of Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaomin Ding
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junyi Yin
- Oncology Department of Tongji Hospital of Tongji University, No. 389 Xincun Road, Shanghai, 200065, China
| | - Xianting Ding
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Haiyan Hu
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hui Yu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
4
|
Yang S, Zou Q, Liang Y, Zhang D, Peng L, Li W, Li W, Liu M, Tong Y, Chen L, Xu P, Yang Z, Zhou K, Xiao J, Wang H, Yu W. miR-1246 promotes osteosarcoma cell migration via NamiRNA-enhancer network dependent on Argonaute 2. MedComm (Beijing) 2024; 5:e543. [PMID: 38585233 PMCID: PMC10999177 DOI: 10.1002/mco2.543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/09/2024] Open
Abstract
High metastatic propensity of osteosarcoma leads to its therapeutic failure and poor prognosis. Although nuclear activation miRNAs (NamiRNAs) are reported to activate gene transcription via targeting enhancer and further promote tumor metastasis, it remains uncertain whether NamiRNAs regulate osteosarcoma metastasis and their exact mechanism. Here, we found that extracellular vesicles of the malignant osteosarcoma cells (143B) remarkably increased the migratory abilities of MNNG cells representing the benign osteosarcoma cells by two folds, which attributed to their high miR-1246 levels. Specially, miR-1246 located in nucleus could activate the migration gene expression (such as MMP1) to accelerate MNNG cell migration through elevating the enhancer activities via increasing H3K27ac enrichment. Instead, MMP1 expression was dramatically inhibited after Argonaute 2 (AGO2) knockdown. Notably, in vitro assays demonstrated that AGO2 recognized the hybrids of miR-1246 and its enhancer DNA via PAZ domains to prevent their degradation from RNase H and these protective roles of AGO2 may favor the gene activation by miR-1246 in vivo. Collectively, our findings suggest that miR-1246 could facilitate osteosarcoma metastasis through interacting with enhancer to activate gene expression dependent on AGO2, highlighting the nuclear AGO2 as a guardian for NamiRNA-targeted gene activation and the potential of miR-1246 for osteosarcoma metastasis therapy.
Collapse
Affiliation(s)
- Shuai Yang
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qingping Zou
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ying Liang
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Dapeng Zhang
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Centre for Eco‐Environmental SciencesChinese Academy of SciencesBeijingChina
| | - Lina Peng
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wei Li
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wenxuan Li
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Mengxing Liu
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ying Tong
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Lu Chen
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Peng Xu
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zhicong Yang
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Kaicheng Zhou
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jianru Xiao
- Department of Orthopaedic OncologyChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Centre for Eco‐Environmental SciencesChinese Academy of SciencesBeijingChina
| | - Wenqiang Yu
- Shanghai Public Health Clinical Centre and Department of General SurgeryHuashan HospitalCancer Metastasis Institute and Laboratory of RNA EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
5
|
Tahara S, Sharma S, de Faria FCC, Sarchet P, Tomasello L, Rentsch S, Karna R, Calore F, Pollock RE. Comparison of three-dimensional cell culture techniques of dedifferentiated liposarcoma and their integration with future research. Front Cell Dev Biol 2024; 12:1362696. [PMID: 38500686 PMCID: PMC10945377 DOI: 10.3389/fcell.2024.1362696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
Background: Dedifferentiated liposarcoma is a formidable sarcoma subtype due to its high local recurrence rate and resistance to medical treatment. While 2D cell cultures are still commonly used, 3D cell culture systems have emerged as a promising alternative, particularly scaffold-based techniques that enable the creation of 3D models with more accurate cell-stroma interactions. Objective: To investigate how 3D structures with or without the scaffold existence would affect liposarcoma cell lines growth morphologically and biologically. Methods: Lipo246 and Lipo863 cell lines were cultured in 3D using four different methods; Matrigel® ECM scaffold method, Collagen ECM scaffold method, ULA plate method and Hanging drop method, in addition to conventional 2D cell culture methods. All samples were processed for histopathological analysis (HE, IHC and DNAscope™), Western blot, and qPCR; moreover, 3D collagen-based models were treated with different doses of SAR405838, a well-known inhibitor of MDM2, and cell viability was assessed in comparison to 2D model drug response. Results: Regarding morphology, cell lines behaved differently comparing the scaffold-based and scaffold-free methods. Lipo863 formed spheroids in Matrigel® but not in collagen, while Lipo246 did not form spheroids in either collagen or Matrigel®. On the other hand, both cell lines formed spheroids using scaffold-free methods. All samples retained liposarcoma characteristic, such as high level of MDM2 protein expression and MDM2 DNA amplification after being cultivated in 3D. 3D collagen samples showed higher cell viability after SAR40538 treatment than 2D models, while cells sensitive to the drug died by apoptosis or necrosis. Conclusion: Our results prompt us to extend our investigation by applying our 3D models to further oncological relevant applications, which may help address unresolved questions about dedifferentiated liposarcoma biology.
Collapse
Affiliation(s)
- Sayumi Tahara
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Soumya Sharma
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Fernanda Costas Casal de Faria
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Patricia Sarchet
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Luisa Tomasello
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sydney Rentsch
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Roma Karna
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Federica Calore
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Raphael E. Pollock
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
6
|
Deng C, Xu Y, Chen H, Zhu X, Huang L, Chen Z, Xu H, Song G, Lu J, Huang W, Liu R, Tang Q, Wang J. Extracellular-vesicle-packaged S100A11 from osteosarcoma cells mediates lung premetastatic niche formation by recruiting gMDSCs. Cell Rep 2024; 43:113751. [PMID: 38341855 DOI: 10.1016/j.celrep.2024.113751] [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: 05/16/2023] [Revised: 11/28/2023] [Accepted: 01/22/2024] [Indexed: 02/13/2024] Open
Abstract
The premetastatic niche (PMN) contributes to lung-specific metastatic tropism in osteosarcoma. However, the crosstalk between primary tumor cells and lung stromal cells is not clearly defined. Here, we dissect the composition of immune cells in the lung PMN and identify granulocytic myeloid-derived suppressor cell (gMDSC) infiltration as positively associated with immunosuppressive PMN formation and tumor cell colonization. Osteosarcoma-cell-derived extracellular vesicles (EVs) activate lung interstitial macrophages to initiate the influx of gMDSCs via secretion of the chemokine CXCL2. Proteomic profiling of EVs reveals that EV-packaged S100A11 stimulates the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway in macrophages by interacting with USP9X. High level of S100A11 expression or circulating gMDSCs correlates with the presentation of lung metastasis and poor prognosis in osteosarcoma patients. In summary, we identify a key role of tumor-derived EVs in lung PMN formation, providing potential strategies for monitoring or preventing lung metastasis in osteosarcoma.
Collapse
Affiliation(s)
- Chuangzhong Deng
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Yanyang Xu
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Hongmin Chen
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Xiaojun Zhu
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Lihua Huang
- State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Administration Department of Nosocomial Infection, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Zhihao Chen
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Huaiyuan Xu
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Guohui Song
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Wenlin Huang
- State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Ranyi Liu
- State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Jin Wang
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; State Key Laboratory of Oncology in Southern China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|
7
|
Wong C, Stoilova I, Gazeau F, Herbeuval JP, Fourniols T. Mesenchymal stromal cell derived extracellular vesicles as a therapeutic tool: immune regulation, MSC priming, and applications to SLE. Front Immunol 2024; 15:1355845. [PMID: 38390327 PMCID: PMC10881725 DOI: 10.3389/fimmu.2024.1355845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by a dysfunction of the immune system. Mesenchymal stromal cell (MSCs) derived extracellular vesicles (EVs) are nanometer-sized particles carrying a diverse range of bioactive molecules, such as proteins, miRNAs, and lipids. Despite the methodological disparities, recent works on MSC-EVs have highlighted their broad immunosuppressive effect, thus driving forwards the potential of MSC-EVs in the treatment of chronic diseases. Nonetheless, their mechanism of action is still unclear, and better understanding is needed for clinical application. Therefore, we describe in this review the diverse range of bioactive molecules mediating their immunomodulatory effect, the techniques and possibilities for enhancing their immune activity, and finally the potential application to SLE.
Collapse
Affiliation(s)
- Christophe Wong
- EVerZom, Paris, France
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | - Ivana Stoilova
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | - Florence Gazeau
- Matière et Systèmes Complexes (MSC) UMR CNRS 7057, Université Paris Cité, Paris, France
| | - Jean-Philippe Herbeuval
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8601, Université Paris Cité, Paris, France
- Chemistry and Biology, Modeling and Immunology for Therapy (CBMIT), Université Paris Cité, Paris, France
| | | |
Collapse
|
8
|
Al-Ansari N, Samuel SM, Büsselberg D. Unveiling the Protective Role of Melatonin in Osteosarcoma: Current Knowledge and Limitations. Biomolecules 2024; 14:145. [PMID: 38397382 PMCID: PMC10886489 DOI: 10.3390/biom14020145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Melatonin, an endogenous neurohormone produced by the pineal gland, has received increased interest due to its potential anti-cancer properties. Apart from its well-known role in the sleep-wake cycle, extensive scientific evidence has shown its role in various physiological and pathological processes, such as inflammation. Additionally, melatonin has demonstrated promising potential as an anti-cancer agent as its function includes inhibition of tumorigenesis, induction of apoptosis, and regulation of anti-tumor immune response. Although a precise pathophysiological mechanism is yet to be established, several pathways related to the regulation of cell cycle progression, DNA repair mechanisms, and antioxidant activity have been implicated in the anti-neoplastic potential of melatonin. In the current manuscript, we focus on the potential anti-cancer properties of melatonin and its use in treating and managing pediatric osteosarcoma. This aggressive bone tumor primarily affects children and adolescents and is treated mainly by surgical and radio-oncological interventions, which has improved survival rates among affected individuals. Significant disadvantages to these interventions include disease recurrence, therapy-related toxicity, and severe/debilitating side effects that the patients have to endure, significantly affecting their quality of life. Melatonin has therapeutic effects when used for treating osteosarcoma, attributed to its ability to halt cancer cell proliferation and trigger apoptotic cell death, thereby enhancing chemotherapeutic efficacy. Furthermore, the antioxidative function of melatonin alleviates harmful side effects of chemotherapy-induced oxidative damage, aiding in decreasing therapeutic toxicities. The review concisely explains the many mechanisms by which melatonin targets osteosarcoma, as evidenced by significant results from several in vitro and animal models. Nevertheless, if further explored, human trials remain a challenge that could shed light and support its utility as an adjunctive therapeutic modality for treating osteosarcoma.
Collapse
Affiliation(s)
- Nojoud Al-Ansari
- Department of Medical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar;
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
| |
Collapse
|
9
|
Molecular and Translational Research on Bone Tumors. Int J Mol Sci 2023; 24:ijms24031946. [PMID: 36768270 PMCID: PMC9916411 DOI: 10.3390/ijms24031946] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Primary bone tumors (PBTs) represent a huge variety of rare malignancies that originate in the skeletal system [...].
Collapse
|
10
|
Zhang J, Li H. Identification of potential extracellular vesicle protein markers altered in osteosarcoma from public databases. Proteomics Clin Appl 2022:e2200084. [PMID: 36571514 DOI: 10.1002/prca.202200084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 12/27/2022]
Abstract
PURPOSE Extracellular vesicles (EVs) have become promising biomarkers for cancer management. Particularly, the molecular cargo such as proteins carried by EVs are similar to their cells of origin, providing important information that can be used for cancer diagnostics, prognosis, and treatment monitoring. However, to date, molecular analysis on EVs is still challenging, limited by the availability of efficient analytical technologies, largely due to the small size of EVs. In this work, we developed a computational workflow for in silico identification of potential EV protein markers from genomic and proteomic databases, and applied it for the discovery of osteosarcoma (OS) EV protein markers. EXPERIMENTAL DESIGN Both mRNA and protein data were computed and compared from publicly accessible databases, and top markers with high differential expression levels were selected. RESULTS Thirty nine markers were identified overexpressed and seven found to be downregulated. These identified markers have been found to be associated with OS on different aspects in literature, demonstrating the usability of this workflow. CONCLUSIONS AND CLINICAL RELEVANCE This work provides a list of potential EV protein markers that are either overexpressed or downregulated in OS for further experimental validation for improved clinical management of OS.
Collapse
Affiliation(s)
- Jinhe Zhang
- School of Engineering, University of Guelph, Guelph, Ontario, Canada
| | - Huiyan Li
- School of Engineering, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
11
|
Gao X, Gao B, Li S. Extracellular vesicles: A new diagnostic biomarker and targeted drug in osteosarcoma. Front Immunol 2022; 13:1002742. [PMID: 36211364 PMCID: PMC9539319 DOI: 10.3389/fimmu.2022.1002742] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma (OS) is a primary bone cancer that is highly prevalent among adolescents and adults below the age of 20 years. The prognostic outcome of metastatic OS or relapse is extremely poor; thus, developing new diagnostic and therapeutic strategies for treating OS is necessary. Extracellular vesicles (EVs) ranging from 30–150 nm in diameter are commonly produced in different cells and are found in various types of body fluids. EVs are rich in biologically active components like proteins, lipids, and nucleic acids. They also strongly affect pathophysiological processes by modulating the intercellular signaling pathways and the exchange of biomolecules. Many studies have found that EVs influence the occurrence, development, and metastasis of osteosarcoma. The regulation of inflammatory communication pathways by EVs affects OS and other bone-related pathological conditions, such as osteoarthritis and rheumatoid arthritis. In this study, we reviewed the latest findings related to diagnosis, prognosis prediction, and the development of treatment strategies for OS from the perspective of EVs.
Collapse
Affiliation(s)
- Xiaozhuo Gao
- Department of Pathology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
| | - Bo Gao
- Department of Pathology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
- *Correspondence: Shenglong Li, ;
| |
Collapse
|
12
|
Keremu A, Aila P, Tusun A, Abulikemu M, Zou X. Extracellular vesicles from bone mesenchymal stem cells transport microRNA-206 into osteosarcoma cells and target NRSN2 to block the ERK1/2-Bcl-xL signaling pathway. Eur J Histochem 2022; 66. [PMID: 35730574 PMCID: PMC9251612 DOI: 10.4081/ejh.2022.3394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
Osteosarcoma (OS) is a kind of malignant tumor originating from mesenchymal tissues. Bone mesenchymal stem cells-derived extracellular vesicles (BMSCs-EVs) can play important roles in OS. This study investigated the mechanism of BMSCs-EVs on OS. BMSC surface antigens and adipogenic and osteogenic differentiation were detected by flow cytometry, and oil red O and alizarin red staining. EVs were isolated from BMSCs by differential centrifugation and identified by transmission electron microscopy, nanoparticle tracking analysis, and Western blot (WB). miR-206 and neurensin-2 (NRSN2) levels in human osteoblast hFOB 1.19 or OS cells (143B, MG-63, Saos2, HOS) were detected by RT-qPCR. Human OS cells with lower miR-206 levels were selected and treated with BMSCs-EVs or pSUPER-NRSN2. The uptake of EVs by 143B cells, cell proliferation, apoptosis, invasion, and migration were detected by immunofluorescence, 5-ethynyl-2’-deoxyuridine (EdU) and colony formation assays, flow cytometry, scratch test, and transwell assays. The binding sites between miR-206 and NRSN2 were predicted by Starbase database and verified by dual-luciferase assay. The OS xenograft model was established and treated with BMSCs-EVs. Tumor growth rate and volume, cell proliferation, and p-ERK1/2, ERK1/2, and Bcl-xL levels were detected by vernier caliper, immunohistochemistry, and WB. BMSCs-EVs were successfully extracted. miR-206 was diminished and NRSN2 was promoted in OS cells. BMSCs-EVs inhibited proliferation, migration, and invasion, and promoted apoptosis of OS cells. BMSCs-EVs carried miR-206 into OS cells. Inhibition of miR-206 in EVs partially reversed the inhibitory effect of EVs on malignant behaviors of OS cells. miR-206 targeted NRSN2. Overexpression of NRSN2 reversed the inhibitory effect of EVs on OS cells. NRSN2 activated the ERK1/2-Bcl-xL pathway. BMSC-EVs inhibited OS growth in vivo. In summary, BMSC-EVs targeted NRSN2 and inhibited the ERK1/2-Bcl-xL pathway by carrying miR-206 into OS cells, thus inhibiting OS progression.
Collapse
Affiliation(s)
- Alimu Keremu
- Orthopedic Center, First People's Hospital of Kashgar, Xinjiang.
| | - Pazila Aila
- Orthopedic Center, First People's Hospital of Kashgar, Xinjiang.
| | - Aikebaier Tusun
- Orthopedic Center, First People's Hospital of Kashgar, Xinjiang.
| | | | - Xiaoguang Zou
- Orthopedic Center, First People's Hospital of Kashgar, Xinjiang.
| |
Collapse
|
13
|
Kumar P, Zadjali F, Yao Y, Köttgen M, Hofherr A, Gross KW, Mehta D, Bissler JJ. Single Gene Mutations in Pkd1 or Tsc2 Alter Extracellular Vesicle Production and Trafficking. BIOLOGY 2022; 11:biology11050709. [PMID: 35625437 PMCID: PMC9139108 DOI: 10.3390/biology11050709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/20/2022] [Accepted: 04/30/2022] [Indexed: 12/17/2022]
Abstract
Simple Summary Extracellular vesicles shed from primary cilia may be involved in renal cystogenesis. The disruption of the Pkd1 gene in our cell culture system increased the production of EVs in a similar way that occurs when the Tsc2 gene is disrupted. Disruption of the primary cilia depresses EV production, and this may be the reason that the combined Kif3A/Pkd1 mutant mouse has a less severe phenotype than the Pkd1 mutant alone. We initiated studies aimed at understanding the renal trafficking of renally-derived EVs and found that single gene disruptions can alter the EV kinetics based on dye tracking studies. These results raise the possibility that EV features, such as cargo, dose, tissue half-life, and targeting, may be involved in the disease process, and these features may also be fertile targets for diagnostic, prognostic, and therapeutic investigation. Abstract Patients with autosomal dominant polycystic kidney disease (ADPKD) and tuberous sclerosis complex (TSC) are born with normal or near-normal kidneys that later develop cysts and prematurely lose function. Both renal cystic diseases appear to be mediated, at least in part, by disease-promoting extracellular vesicles (EVs) that induce genetically intact cells to participate in the renal disease process. We used centrifugation and size exclusion chromatography to isolate the EVs for study. We characterized the EVs using tunable resistive pulse sensing, dynamic light scattering, transmission electron microscopy, and Western blot analysis. We performed EV trafficking studies using a dye approach in both tissue culture and in vivo studies. We have previously reported that loss of the Tsc2 gene significantly increased EV production and here demonstrate that the loss of the Pkd1 gene also significantly increases EV production. Using a cell culture system, we also show that loss of either the Tsc2 or Pkd1 gene results in EVs that exhibit an enhanced uptake by renal epithelial cells and a prolonged half-life. Loss of the primary cilia significantly reduces EV production in renal collecting duct cells. Cells that have a disrupted Pkd1 gene produce EVs that have altered kinetics and a prolonged half-life, possibly impacting the duration of the EV cargo effect on the recipient cell. These results demonstrate the interplay between primary cilia and EVs and support a role for EVs in polycystic kidney disease pathogenesis.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- US FDA National Center for Toxicological Research, Jefferson, AR 72079, USA;
| | - Fahad Zadjali
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Ying Yao
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | - Michael Köttgen
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (M.K.); (A.H.)
- CIBSS—Centre for Integrative Biological Signaling Studies, 79104 Freiburg, Germany
| | - Alexis Hofherr
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (M.K.); (A.H.)
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Darshan Mehta
- US FDA National Center for Toxicological Research, Jefferson, AR 72079, USA;
| | - John J. Bissler
- Department of Pediatrics, Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (P.K.); (F.Z.); (Y.Y.)
- Children’s Foundation Research Institute (CFRI), Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- Pediatric Medicine Department, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Correspondence:
| |
Collapse
|
14
|
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
|
15
|
Li M, Yin H, Yan Z, Li H, Wu J, Wang Y, Wei F, Tian G, Ning C, Li H, Gao C, Fu L, Jiang S, Chen M, Sui X, Liu S, Chen Z, Guo Q. The immune microenvironment in cartilage injury and repair. Acta Biomater 2022; 140:23-42. [PMID: 34896634 DOI: 10.1016/j.actbio.2021.12.006] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023]
Abstract
The ability of articular cartilage to repair itself is limited because it lacks blood vessels, nerves, and lymph tissue. Once damaged, it can lead to joint swelling and pain, accelerating the progression of osteoarthritis. To date, complete regeneration of hyaline cartilage exhibiting mechanical properties remains an elusive goal, despite the many available technologies. The inflammatory milieu created by cartilage damage is critical for chondrocyte death and hypertrophy, extracellular matrix breakdown, ectopic bone formation, and progression of cartilage injury to osteoarthritis. In the inflammatory microenvironment, mesenchymal stem cells (MSCs) undergo aberrant differentiation, and chondrocytes begin to convert or dedifferentiate into cells with a fibroblast phenotype, thereby resulting in fibrocartilage with poor mechanical qualities. All these factors suggest that inflammatory problems may be a major stumbling block to cartilage repair. To produce a milieu conducive to cartilage repair, multi-dimensional management of the joint inflammatory microenvironment in place and time is required. Therefore, this calls for elucidation of the immune microenvironment of cartilage repair after injury. This review provides a brief overview of: (1) the pathogenesis of cartilage injury; (2) immune cells in cartilage injury and repair; (3) effects of inflammatory cytokines on cartilage repair; (4) clinical strategies for treating cartilage defects; and (5) strategies for targeted immunoregulation in cartilage repair. STATEMENT OF SIGNIFICANCE: Immune response is increasingly considered the key factor affecting cartilage repair. It has both negative and positive regulatory effects on the process of regeneration and repair. Proinflammatory factors are secreted in large numbers, and necrotic cartilage is removed. During the repair period, immune cells can secrete anti-inflammatory factors and chondrogenic cytokines, which can inhibit inflammation and promote cartilage repair. However, inflammatory factors persist, which accelerate the degradation of the cartilage matrix. Furthermore, in an inflammatory microenvironment, MSCs undergo abnormal differentiation, and chondrocytes begin to transform or dedifferentiate into fibroblast-like cells, forming fibrocartilage with poor mechanical properties. Consequently, cartilage regeneration requires multi-dimensional regulation of the joint inflammatory microenvironment in space and time to make it conducive to cartilage regeneration.
Collapse
|
16
|
FT-IR Spectral Signature of Sensitive and Multidrug-Resistant Osteosarcoma Cell-Derived Extracellular Nanovesicles. Cells 2022; 11:cells11050778. [PMID: 35269400 PMCID: PMC8909163 DOI: 10.3390/cells11050778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone cancer in children and adolescents. Despite aggressive treatment regimens, the outcome is unsatisfactory, and multidrug resistance (MDR) is a pivotal process in OS treatment failure. OS-derived extracellular vesicles (EVs) promote drug resistance to chemotherapy and target therapy through different mechanisms. The aim of this study was to identify subpopulations of osteosarcoma-EVs by Fourier transform infrared spectroscopy (FT-IR) to define a specific spectral signature for sensitive and multidrug-resistant OS-derived EVs. EVs were isolated from sensitive and MDR OS cells as well as from mesenchymal stem cells by differential centrifugation and ultracentrifugation. EVs size, morphology and protein expression were characterized. FT-IR/ATR of EVs spectra were acquired in the region of 400–4000 cm−1 (resolution 4 cm−1, 128 scans). The FT-IR spectra obtained were consistently different in the EVs compared to cells from which they originate. A specific spectral signature, characterized by a shift and a new band (1601 cm−1), permitted to clearly distinguish EVs isolated by sensitive and multidrug-resistant OS cells. Our data suggest that FT-IR spectroscopy allows to characterize and define a specific spectral signature for sensitive and MDR OS-derived EVs.
Collapse
|
17
|
Yang Q, Liu J, Wu B, Wang X, Jiang Y, Zhu D. Role of extracellular vesicles in osteosarcoma. Int J Med Sci 2022; 19:1216-1226. [PMID: 35928720 PMCID: PMC9346389 DOI: 10.7150/ijms.74137] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Osteosarcoma is a malignant bone tumor characterized by the direct production of osteoid tissue from tumor cells. Extracellular vesicles are membranous vesicles released by cells into the extracellular matrix, which exist widely in various body fluids and cell supernatants, and stably carry some important signaling molecules. They are involved in cell communication, cell migration, angiogenesis and tumor cell growth. Increasing evidence has shown that extracellular vesicles play a significant role in osteosarcoma development, progression, and metastatic process, indicating that extracellular vesicles can be use as biomarker vehicles in the diagnosis and prognosis of osteosarcoma. This review discusses the basic biological characteristics of extracellular vesicles and focuses on their application in osteosarcoma.
Collapse
Affiliation(s)
- Qifan Yang
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Jing Liu
- The first clinical medical college of Bin Zhou Medical College, Street Huanghe 661, China
| | - Bo Wu
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Xinyu Wang
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Ye Jiang
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| | - Dong Zhu
- Department of Orthopaedics, the First Hospital of Jilin University, Changchun, Street Xinmin 71, China
| |
Collapse
|
18
|
Advanced approaches to regenerate spinal cord injury: The development of cell and tissue engineering therapy and combinational treatments. Biomed Pharmacother 2021; 146:112529. [PMID: 34906773 DOI: 10.1016/j.biopha.2021.112529] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is a central nervous system (CNS) devastate event that is commonly caused by traumatic or non-traumatic events. The reinnervation of spinal cord axons is hampered through a myriad of devices counting on the damaged myelin, inflammation, glial scar, and defective inhibitory molecules. Unfortunately, an effective treatment to completely repair SCI and improve functional recovery has not been found. In this regard, strategies such as using cells, biomaterials, biomolecules, and drugs have been reported to be effective for SCI recovery. Furthermore, recent advances in combinatorial treatments, which address various aspects of SCI pathophysiology, provide optimistic outcomes for spinal cord regeneration. According to the global importance of SCI, the goal of this article review is to provide an overview of the pathophysiology of SCI, with an emphasis on the latest modes of intervention and current advanced approaches for the treatment of SCI, in conjunction with an assessment of combinatorial approaches in preclinical and clinical trials. So, this article can give scientists and clinicians' clues to help them better understand how to construct preclinical and clinical studies that could lead to a breakthrough in spinal cord regeneration.
Collapse
|
19
|
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: 2.8] [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
|
20
|
Li S. The basic characteristics of extracellular vesicles and their potential application in bone sarcomas. J Nanobiotechnology 2021; 19:277. [PMID: 34535153 PMCID: PMC8447529 DOI: 10.1186/s12951-021-01028-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
Bone sarcomas are rare cancers accompanied by metastatic disease, mainly including osteosarcoma, Ewing sarcoma and chondrosarcoma. Extracellular vesicles (EVs) are membrane vesicles released by cells in the extracellular matrix, which carry important signal molecules, can stably and widely present in various body fluids, such as plasma, saliva and scalp fluid, spinal cord, breast milk, and urine liquid. EVs can transport almost all types of biologically active molecules (DNA, mRNA, microRNA (miRNA), proteins, metabolites, and even pharmacological compounds). In this review, we summarized the basic biological characteristics of EVs and focused on their application in bone sarcomas. EVs can be use as biomarker vehicles for diagnosis and prognosis in bone sarcomas. The role of EVs in bone sarcoma has been analyzed point-by-point. In the microenvironment of bone sarcoma, bone sarcoma cells, mesenchymal stem cells, immune cells, fibroblasts, osteoclasts, osteoblasts, and endothelial cells coexist and interact with each other. EVs play an important role in the communication between cells. Based on multiple functions in bone sarcoma, this review provides new ideas for the discovery of new therapeutic targets and new diagnostic analysis.
Collapse
Affiliation(s)
- Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning Province, China.
- Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| |
Collapse
|
21
|
Chang X, Ma Z, Zhu G, Lu Y, Yang J. New perspective into mesenchymal stem cells: Molecular mechanisms regulating osteosarcoma. J Bone Oncol 2021; 29:100372. [PMID: 34258182 PMCID: PMC8254115 DOI: 10.1016/j.jbo.2021.100372] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/14/2021] [Accepted: 06/02/2021] [Indexed: 02/05/2023] Open
Abstract
The origin of osteosarcoma cells from osteoblasts and mesenchymal stem cells remains controversial. Mesenchymal stem cells regulate the development of osteosarcoma by influencing the tumor microenvironment and mediating cell communication. Mesenchymal stem cells and exosomes secreted by them can be used as good genes and drug carriers for the treatment of osteosarcoma. Mesenchymal stem cells from different tissue sources have different regulatory effects on the development of osteosarcoma.
Mesenchymal stem cells (MSCs) are multipotent stem cells with significant potential for regenerative medicine. The tumorigenesis of osteosarcoma is an intricate system and MSCs act as an indispensable part of this, interacting with the tumor microenvironment (TME) during the process. MSCs link to cells by acting on each component in the TME via autocrine or paracrine extracellular vesicles for cellular communication. Because of their unique characteristics, MSCs can be modified and processed into good biological carriers, loaded with drugs, and transfected with anticancer genes for the targeted treatment of osteosarcoma. Previous high-quality reviews have described the biological characteristics of MSCs; this review will discuss the effects of MSCs on the components of the TME and cellular communication and the prospects for clinical applications of MSCs.
Collapse
Key Words
- 3TSR, Three type 1 repeats
- 5 FC, 5-fluorocytosine
- AD-MSCs, Adipose-derived MSCs
- AQP1, Aquaporin-1
- BMSC-derived exosomes, BMSC-Exos
- BMSCs, Bone marrow mesenchymal stem cells
- CAFs, Carcinoma-associated-fibroblasts
- CRC, Colorectal cancer
- CSF, Colony-stimulating factor
- Cellular communication
- Clinical application
- DOX, Doxorubicin
- DP-MSCs, Dental pulp-derived MSCs, hUC-MSCs, Human umbilical cord MSCs
- ECM, Extracellular matrix
- ESCs, embryonic stem cells
- EVs, Extracellular vesicles
- GBM, Glioblastoma
- HCC, hepatocellular carcinoma
- LINE-1, Long interspersing element 1
- MCP-1, Monocyte chemoattractant protein-1
- MSC-Exos, MSC-derived exosomes
- MSC-MVs, MSC microvesicles
- MSCs
- MSCs, Mesenchymal stem cells
- OPG, osteoprotegerin
- OS, osteosarcoma
- Osteosarcoma
- PDGFRα, Platelet derived growth factor receptor α
- PDGFRβ, Platelet derived growth factor receptor β
- PDGFα, Platelet derived growth factor α
- S TRAIL, Secretable variant of the TNF-related apoptosis-inducing ligand
- SD-MSCs, stressed MSCs
- SDF-1, Stromal cell-derived factor 1
- TGF, Transforming growth factor
- TME
- TME, Tumor microenvironment
- TNF, Tumor necrosis factor
- TRA2B, Transformer 2β
- VEGF, Vascular endothelial growth factor
- hASCs, human adipose stem cells
- iPSCs, induced pluripotent stem cells
- yCD::UPRT, Yeast cytosine deaminase::uracil phosphoribosyl transferase
Collapse
Affiliation(s)
- Xingyu Chang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zhanjun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Guomao Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yubao Lu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jingjing Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
22
|
Quiroz-Acosta T, Flores-Martinez YM, Becerra-Martínez E, Pérez-Hernández E, Pérez-Hernández N, Bañuelos-Hernández AE. Aberrant sphingomyelin 31P-NMR signatures in giant cell tumour of bone. Biochem Cell Biol 2021; 99:717-724. [PMID: 34096319 DOI: 10.1139/bcb-2020-0599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An understanding of the biochemistry of the giant cell tumour of bone (GCTB) provides an opportunity for the development of prognostic markers and identification of therapeutic targets. Based on metabolomic analysis, we proposed glycerophospholipid metabolism as the altered pathway in GCTB and the objective of this study was to identify these altered metabolites. Using phosphorus-31 nuclear magnetic resonance spectroscopy (31P-NMR), sphingomyelin was determined as the most dysregulated phospholipid in tissue samples from six patients with GCTB; subsequently, enzymes related to its biosynthesis and hydrolysis were examined using immunodetection techniques. High expression of sphingomyelin synthases 1 and 2, but low expression of neutral sphingomyelinase 2 (nSMase2), was found in GCTB tissues compared to non-neoplastic bone tissues. Sphingomyelin/ ceramide biosynthesis is dysregulated in GCTB due to alterations in the expression of SMS1, SMS2, and nSMase2.
Collapse
Affiliation(s)
- Tayde Quiroz-Acosta
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Mexico, Ciudad de México, Mexico;
| | - Yazmin Montserrat Flores-Martinez
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Mexico, Ciudad de México, Mexico;
| | - Elvia Becerra-Martínez
- Centro de Nanociencias y Micro y Nanotecnologías, Instituto Politécnico Nacional, México, Ciudad de México, Mexico;
| | - Elizabeth Pérez-Hernández
- UMAE de Traumatología, Ortopedia y Rehabilitación "Dr. Victorio de la Fuente Narváez", Mexico, Ciudad de México, Mexico;
| | - Nury Pérez-Hernández
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Mexico, Ciudad de México, Mexico;
| | - Angel Ernesto Bañuelos-Hernández
- Centro de Investigacion y de Estudios Avanzados del Instituto Politecnico Nacional, 42576, Departamento de Farmacologia, Ciudad de Mexico, Mexico City, Mexico;
| |
Collapse
|
23
|
MAGEA4 Coated Extracellular Vesicles Are Stable and Can Be Assembled In Vitro. Int J Mol Sci 2021; 22:ijms22105208. [PMID: 34069064 PMCID: PMC8155938 DOI: 10.3390/ijms22105208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/02/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) are valued candidates for the development of new tools for medical applications. Vesicles carrying melanoma-associated antigen A (MAGEA) proteins, a subfamily of cancer-testis antigens, are particularly promising tools in the fight against cancer. Here, we have studied the biophysical and chemical properties of MAGEA4-EVs and show that they are stable under common storage conditions such as keeping at +4 °C and -80 °C for at least 3 weeks after purification. The MAGEA4-EVs can be freeze-thawed two times without losing MAGEA4 in detectable quantities. The attachment of MAGEA4 to the surface of EVs cannot be disrupted by high salt concentrations or chelators, but the vesicles are sensitive to high pH. The MAGEA4 protein can bind to the surface of EVs in vitro, using robust passive incubation. In addition, EVs can be loaded with recombinant proteins fused to the MAGEA4 open reading frame within the cells and also in vitro. The high stability of MAGEA4-EVs ensures their potential for the development of EV-based anti-cancer applications.
Collapse
|
24
|
Edmunds GL, Smalley MJ, Beck S, Errington RJ, Gould S, Winter H, Brodbelt DC, O'Neill DG. Dog breeds and body conformations with predisposition to osteosarcoma in the UK: a case-control study. Canine Med Genet 2021; 8:2. [PMID: 33750475 PMCID: PMC7944903 DOI: 10.1186/s40575-021-00100-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/30/2021] [Indexed: 11/16/2022] Open
Abstract
Background Osteosarcoma is an aggressive and painful bone neoplasm in dogs. Previous studies have reported epidemiological associations suggesting that large body mass, long bone length and the genetics of certain breeds including the Rottweiler are associated with elevated osteosarcoma risk. However, these studies were often limited by selection bias and confounding factors, and have rarely offered insights into breed-associated protection for osteosarcoma. The current study includes 1756 appendicular and axial osteosarcoma cases presenting to VPG Histology (Bristol, UK) compared against a control population of 905,211 dogs without osteosarcoma from primary care electronic patient records in the VetCompass™ dataset. Methods and study design Retrospective, case-control study. Multivariable logistic regression analysis explored associations between demographic risk factors (including breed, chondrodystrophy, age, sex/neuter status, skull-shape, and body mass) and osteosarcoma of all anatomical sites. Results We identified several breeds with increased and reduced odds of osteosarcoma. At highest risk were the Rottweiler and Great Dane, with > 10 times the odds of osteosarcoma compared with crossbreds, and the Rhodesian Ridgeback, which has not featured in previous lists of at-risk breeds for osteosarcoma, and had an odds ratio of 11.31 (95% confidence interval 7.37–17.35). Breeds at lowest risk of osteosarcoma (protected breeds) included the Bichon Frise, the French Bulldog and the Cavalier King Charles Spaniel, all with odd ratios of less than 0.30 compared with crossbreds. Body mass was strongly associated with osteosarcoma risk; dogs over 40 kg exhibited osteosarcoma odds of 45.44 (95% confidence interval 33.74–61.20) compared with dogs less than 10 kg. Chondrodystrophic breeds had an osteosarcoma odds ratio of 0.13 (95% confidence interval 0.11–0.16) compared with non-chondrodystrophic breeds. Conclusions This study provides evidence of strong breed-associated osteosarcoma risk and protection, suggesting a genetic basis for osteosarcoma pathogenesis. It highlights that breeds selected for long legs/large body mass are generally overrepresented amongst at-risk breeds, whilst those selected for short leg length/small body mass are generally protected. These findings could inform genetic studies to identify osteosarcoma risk alleles in canines and humans; as well as increasing awareness amongst veterinarians and owners, resulting in improved breeding practices and clinical management of osteosarcoma in dogs. Supplementary Information The online version contains supplementary material available at 10.1186/s40575-021-00100-7.
Collapse
Affiliation(s)
- Grace L Edmunds
- Bristol Veterinary School, Langford House, Langford, Bristol, BS40 5DU, UK.
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Sam Beck
- VPG Histology (formerly Bridge), Horner Court, 637 Gloucester Road, Horfield, Bristol, BS7 0BJ, UK
| | - Rachel J Errington
- Division of Cancer and Genetics, School of Medicine, Academic Avenue, Cardiff University, Cardiff, CF62 3LF, UK
| | - Sara Gould
- Langford Veterinary Services, Langford House Langford, Bristol, BS40 5DU, UK
| | | | - Dave C Brodbelt
- Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Herts, AL9 7TA, UK
| | - Dan G O'Neill
- Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Herts, AL9 7TA, UK
| |
Collapse
|
25
|
Li F, Chen X, Shang C, Ying Q, Zhou X, Zhu R, Lu H, Hao X, Dong Q, Jiang Z. Bone Marrow Mesenchymal Stem Cells-Derived Extracellular Vesicles Promote Proliferation, Invasion and Migration of Osteosarcoma Cells via the lncRNA MALAT1/miR-143/NRSN2/Wnt/β-Catenin Axis. Onco Targets Ther 2021; 14:737-749. [PMID: 33564242 PMCID: PMC7866913 DOI: 10.2147/ott.s283459] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/25/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Osteosarcoma is a malignant primary bone tumor. Bone marrow-derived mesenchymal stem cells-derived extracellular vesicles (BMSC-EVs) bear repair function for bone and cartilage. This study investigated the mechanism of BMSC-EVs in osteosarcoma cell proliferation, migration and invasion. Methods BMSC-EVs were isolated and identified. The effects of different concentrations of EVs on osteosarcoma cell proliferation, migration and invasion were evaluated. LncRNA MALAT1 expression in osteosarcoma cells was detected. BMSCs were transfected with si-MALAT1 or si-NC. The binding relationships between MALAT1 and miR-143, and miR-143 and NRSN2 were verified. Levels of NRSN2 and Wnt/β-catenin pathway key proteins were detected. miR-143 mimic was transfected into EVs-treated osteosarcoma cells. Nude mice were injected with MG63 cells to verify the effect of EVs on osteosarcoma growth in vivo. Results BMSC-EVs facilitated proliferation, invasion and migration of osteosarcoma cells. BMSC-EVs carried MALAT1 into osteosarcoma cells. BMSC-EVs-treated osteosarcoma cells showed increased MALAT1 and NRSN2 expressions, decreased miR-143 expression, and activated Wnt/β-catenin pathway. miR-143 mimic or si-MALAT1 reversed the effects of BMSC-EVs on osteosarcoma cells. In vivo experiment confirmed that BMSC-EVs promoted tumor growth in nude mice. Discussion BMSC-EVs promoted proliferation, invasion and migration of osteosarcoma cells via the MALAT1/miR-143/NRSN2/Wnt/β-catenin axis. This study might offer new insights into osteosarcoma management.
Collapse
Affiliation(s)
- Fujiang Li
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Xin Chen
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Cong Shang
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Qinglong Ying
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Xianjun Zhou
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Rongkun Zhu
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Hongting Lu
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Xiwei Hao
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Qian Dong
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Zhong Jiang
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| |
Collapse
|
26
|
Perut F, Roncuzzi L, Avnet S, Massa A, Zini N, Sabbadini S, Giampieri F, Mezzetti B, Baldini N. Strawberry-Derived Exosome-Like Nanoparticles Prevent Oxidative Stress in Human Mesenchymal Stromal Cells. Biomolecules 2021; 11:biom11010087. [PMID: 33445656 PMCID: PMC7828105 DOI: 10.3390/biom11010087] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Plant-derived exosome-like nanovesicles (EPDENs) have recently been isolated and evaluated as potential bioactive nutraceutical biomolecules. It has been hypothesized that EPDENs may exert their activity on mammalian cells through their specific cargo. In this study, we isolated and purified EPDENs from the strawberry juice of Fragaria x ananassa (cv. Romina), a new cultivar characterized by a high content of anthocyanins, folic acid, flavonols, and vitamin C and an elevated antioxidant capacity. Fragaria-derived EPDENs were purified by a series of centrifugation and filtration steps. EPDENs showed size and morphology similar to mammalian extracellular nanovesicles. The internalization of Fragaria-derived EPDENs by human mesenchymal stromal cells (MSCs) did not negatively affect their viability, and the pretreatment of MSCs with Fragaria-derived EPDENs prevented oxidative stress in a dose-dependent manner. This is possibly due to the presence of vitamin C inside the nanovesicle membrane. The analysis of EPDEN cargo also revealed the presence of small RNAs and miRNAs. These findings suggest that Fragaria-derived EPDENs may be considered nanoshuttles contained in food, with potential health-promoting activity.
Collapse
Affiliation(s)
- Francesca Perut
- BST Biomedical Sciences and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (F.P.); (L.R.); (S.A.); (A.M.)
| | - Laura Roncuzzi
- BST Biomedical Sciences and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (F.P.); (L.R.); (S.A.); (A.M.)
| | - Sofia Avnet
- BST Biomedical Sciences and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (F.P.); (L.R.); (S.A.); (A.M.)
| | - Annamaria Massa
- BST Biomedical Sciences and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (F.P.); (L.R.); (S.A.); (A.M.)
| | - Nicoletta Zini
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, 40100 Bologna, Italy;
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Silvia Sabbadini
- Department of Agricultural, Food and Environmental Sciences, Università Politecnica delle Marche, 60121 Ancona, Italy; (S.S.); (B.M.)
| | - Francesca Giampieri
- Department of Clinical Specialistic and Odontostomatological Sciences, University Politecnica delle Marche, 60121 Ancona, Italy;
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Bruno Mezzetti
- Department of Agricultural, Food and Environmental Sciences, Università Politecnica delle Marche, 60121 Ancona, Italy; (S.S.); (B.M.)
| | - Nicola Baldini
- BST Biomedical Sciences and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (F.P.); (L.R.); (S.A.); (A.M.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
- Correspondence: ; Tel.: +39-051-6366566
| |
Collapse
|
27
|
Prudowsky ZD, Yustein JT. Recent Insights into Therapy Resistance in Osteosarcoma. Cancers (Basel) 2020; 13:E83. [PMID: 33396725 PMCID: PMC7795058 DOI: 10.3390/cancers13010083] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/22/2022] Open
Abstract
Osteosarcoma, the most common bone malignancy of childhood, has been a challenge to treat and cure. Standard chemotherapy regimens work well for many patients, but there remain minimal options for patients with progressive or resistant disease, as clinical trials over recent decades have failed to significantly improve survival. A better understanding of therapy resistance is necessary to improve current treatments and design new strategies for future treatment options. In this review, we discuss known mechanisms and recent scientific advancements regarding osteosarcoma and its patterns of resistance against chemotherapy, radiation, and other newly-introduced therapeutics.
Collapse
Affiliation(s)
- Zachary D. Prudowsky
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Houston, TX 77030, USA;
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason T. Yustein
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Houston, TX 77030, USA;
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
28
|
Extracellular Vesicles in Hematological Malignancies: From Biomarkers to Therapeutic Tools. Diagnostics (Basel) 2020; 10:diagnostics10121065. [PMID: 33316884 PMCID: PMC7763630 DOI: 10.3390/diagnostics10121065] [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: 11/01/2020] [Revised: 12/06/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023] Open
Abstract
Small extracellular vesicles (EVs) are a heterogenous group of lipid particles released by all cell types in physiological and pathological states. In hematological malignancies, tumor-derived EVs are critical players in mediating intercellular communications through the transfer of genetic materials and proteins between neoplastic cells themselves and to several components of the bone marrow microenvironment, rendering the latter a “stronger” niche supporting cancer cell proliferation, drug resistance, and escape from immune surveillance. In this context, the molecular cargoes of tumor-derived EVs reflect the nature and status of the cells of origin, making them specific therapeutic targets. Another important characteristic of EVs in hematological malignancies is their use as a potential “liquid biopsy” because of their high abundance in biofluids and their ability to protect their molecular cargoes from nuclease and protease degradation. Liquid biopsies are non-invasive blood tests that provide a molecular profiling clinical tool as an alternative method of disease stratification, especially in cancer patients where solid biopsies have limited accessibility. They offer accurate diagnoses and identify specific biomarkers for monitoring of disease progression and response to treatment. In this review, we will focus on the role of EVs in the most prevalent hematological malignancies, particularly on their prospective use as biomarkers in the context of liquid biopsies, as well as their molecular signature that identifies them as specific therapeutic targets for inhibiting cancer progression. We will also highlight their roles in modulating the immune response by acting as both immunosuppressors and activators of anti-tumor immunity.
Collapse
|
29
|
Brain Tumor-Derived Extracellular Vesicles as Carriers of Disease Markers: Molecular Chaperones and MicroRNAs. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10196961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Primary and metastatic brain tumors are usually serious conditions with poor prognosis, which reveal the urgent need of developing rapid diagnostic tools and efficacious treatments. To achieve these objectives, progress must be made in the understanding of brain tumor biology, for example, how they resist natural defenses and therapeutic intervention. One resistance mechanism involves extracellular vesicles that are released by tumors to meet target cells nearby or distant via circulation and reprogram them by introducing their cargo. This consists of different molecules among which are microRNAs (miRNAs) and molecular chaperones, the focus of this article. miRNAs modify target cells in the immune system to avoid antitumor reaction and chaperones are key survival molecules for the tumor cell. Extracellular vesicles cargo reflects the composition and metabolism of the original tumor cell; therefore, it is a source of markers, including the miRNAs and chaperones discussed in this article, with potential diagnostic and prognostic value. This and their relatively easy availability by minimally invasive procedures (e.g., drawing venous blood) illustrate the potential of extracellular vesicles as useful materials to manage brain tumor patients. Furthermore, understanding extracellular vesicles circulation and interaction with target cells will provide the basis for using this vesicle for delivering therapeutic compounds to selected tumor cells.
Collapse
|
30
|
Heymann MF, Lezot F, Heymann D. Bisphosphonates in common pediatric and adult bone sarcomas. Bone 2020; 139:115523. [PMID: 32622877 DOI: 10.1016/j.bone.2020.115523] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 01/23/2023]
Abstract
The therapeutic strategies proposed currently for bone sarcomas are based on neo-adjuvant chemotherapy, delayed en-bloc wide resection, and adjuvant chemotherapy. Unfortunately, bone sarcomas are characterized by high rates of poor drug response, with a high risk of drug resistance, local recurrence and/or a high propensity for induced metastases. The pathogenesis of bone sarcomas is strongly associated with dysregulation of local bone remodeling and increased osteolysis that plays a part in tumor development. In this context, bisphosphonates (BPs) have been proposed as a single agent or in combination with conventional drugs to block bone resorption and the vicious cycle established between bone and sarcoma cells. Pre-clinical in vitro studies revealed the potential "anti-tumor" activities of nitrogen-bisphosphonates (N-BPs). In pre-clinical models, N-BPs reduced significantly primary tumor growth in osteosarcoma and Ewing sarcoma, and the installation of lung metastases. In chondrosarcoma, N-BPs reduced the recurrence of local tumors after intralesional curettage, and increased overall survival. In pediatric and adult osteosarcoma patients, N-BPs have been assessed in combination with conventional chemotherapy and surgery in randomized phase 3 studies with no improvement in clinical outcome. The lack of benefit may potentially be explained by the biological impact of N-BPs on macrophage differentiation/recruitment which may alter CD8+-T lymphocyte infiltration. Thanks to their considerable affinity for the mineralized extracellular matrix, BPs are an excellent platform for drug delivery in malignant bone sites with reduced systemic toxicity, which opens up new opportunities for their future use.
Collapse
Affiliation(s)
- Marie-Francoise Heymann
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France; Université de Nantes, Nantes, France
| | - Frederic Lezot
- Université de Nantes, Inserm, U1238, Faculty of Medicine, Nantes, France
| | - Dominique Heymann
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France; Université de Nantes, Nantes, France; University of Sheffield, Dept of Oncology and Metabolism, School of Medicine, Sheffield, UK.
| |
Collapse
|
31
|
Bone Microenvironment and Osteosarcoma Metastasis. Int J Mol Sci 2020; 21:ijms21196985. [PMID: 32977425 PMCID: PMC7582690 DOI: 10.3390/ijms21196985] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 01/10/2023] Open
Abstract
The bone microenvironment is an ideal fertile soil for both primary and secondary tumors to seed. The occurrence and development of osteosarcoma, as a primary bone tumor, is closely related to the bone microenvironment. Especially, the metastasis of osteosarcoma is the remaining challenge of therapy and poor prognosis. Increasing evidence focuses on the relationship between the bone microenvironment and osteosarcoma metastasis. Many elements exist in the bone microenvironment, such as acids, hypoxia, and chemokines, which have been verified to affect the progression and malignance of osteosarcoma through various signaling pathways. We thoroughly summarized all these regulators in the bone microenvironment and the transmission cascades, accordingly, attempting to furnish hints for inhibiting osteosarcoma metastasis via the amelioration of the bone microenvironment. In addition, analysis of the cross-talk between the bone microenvironment and osteosarcoma will help us to deeply understand the development of osteosarcoma. The cellular and molecular protagonists presented in the bone microenvironment promoting osteosarcoma metastasis will accelerate the exploration of novel therapeutic strategies towards osteosarcoma.
Collapse
|
32
|
Cersosimo F, Lonardi S, Bernardini G, Telfer B, Mandelli GE, Santucci A, Vermi W, Giurisato E. Tumor-Associated Macrophages in Osteosarcoma: From Mechanisms to Therapy. Int J Mol Sci 2020; 21:E5207. [PMID: 32717819 PMCID: PMC7432207 DOI: 10.3390/ijms21155207] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
Osteosarcomas (OSs) are bone tumors most commonly found in pediatric and adolescent patients characterized by high risk of metastatic progression and recurrence after therapy. Effective therapeutic management of this disease still remains elusive as evidenced by poor patient survival rates. To achieve a more effective therapeutic management regimen, and hence patient survival, there is a need to identify more focused targeted therapies for OSs treatment in the clinical setting. The role of the OS tumor stroma microenvironment plays a significant part in the development and dissemination of this disease. Important components, and hence potential targets for treatment, are the tumor-infiltrating macrophages that are known to orchestrate many aspects of OS stromal signaling and disease progression. In particular, increased infiltration of M2-like tumor-associated macrophages (TAMs) has been associated with OS metastasis and poor patient prognosis despite currently used aggressive therapies regimens. This review aims to provide a summary update of current macrophage-centered knowledge and to discuss the possible roles that macrophages play in the process of OS metastasis development focusing on the potential influence of stromal cross-talk signaling between TAMs, cancer-stem cells and additional OSs tumoral microenvironment factors.
Collapse
Affiliation(s)
- Francesca Cersosimo
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (F.C.); (G.B.); (A.S.)
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.L.); (G.E.M.); (W.V.)
| | - Giulia Bernardini
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (F.C.); (G.B.); (A.S.)
| | - Brian Telfer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK;
| | - Giulio Eugenio Mandelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.L.); (G.E.M.); (W.V.)
| | - Annalisa Santucci
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (F.C.); (G.B.); (A.S.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (S.L.); (G.E.M.); (W.V.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (F.C.); (G.B.); (A.S.)
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
33
|
Gu Z, Zhou Y, Cao C, Wang X, Wu L, Ye Z. TFAP2C-mediated LINC00922 signaling underpins doxorubicin-resistant osteosarcoma. Biomed Pharmacother 2020; 129:110363. [PMID: 32563982 DOI: 10.1016/j.biopha.2020.110363] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have been indicated as critical regulators in osteosarcoma (OS). However, the function of lncRNAs in doxorubicin (DXR)-resistant OS remain unclear. Here, present study investigated the functions of lncRNA LINC00922 on the DXR resistance in OS tumorigenesis. LncRNA expression profile was detected using lncRNA microarray in DXR-resistant OS cells (MG63/DXR) and parental cells (MG63). Molecular binding was detected using luciferase reporter assay and chromatin immunoprecipitation. DXR sensitivity assay was detected using CCK-8 assay. Results showed that LINC00922 was significantly up-regulated in OS tissue specimens. Cellular assays showed that LINC00922 increased DXR IC50 and the knockdown of LINC00922 repressed the tumor growth of OS cells. Mechanistic assays showed that LINC00922 acts as a sponge of miR-424-5p, and miR-424-5p targeted the 3'-untranslated region of transcription factor activating protein 2 gamma (TFAP2C) mRNA. Moreover, TFAP2C promoted transcription of LINC00922 in a positive feedback loop comprising TFAP2C, LINC00922, and miR-424-5p. Collectively, these findings uncovered the function of TFAP2C/LINC00922/miR-424-5p feedback loop in DXR resistance, suggesting new therapeutic direction for OS.
Collapse
Affiliation(s)
- Zenghui Gu
- Department of Orthopaedics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Yuanxi Zhou
- Department of Orthopaedics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Chenye Cao
- Department of Orthopaedics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xinqiang Wang
- Department of Orthopaedics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Liangbang Wu
- Department of Orthopaedics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Zhaoming Ye
- Department of Orthopaedics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
34
|
The Role of Extracellular Vesicles in the Hallmarks of Cancer and Drug Resistance. Cells 2020; 9:cells9051141. [PMID: 32384712 PMCID: PMC7290603 DOI: 10.3390/cells9051141] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular signaling and communication, allowing the intercellular exchange of proteins, lipids, and genetic material. Their recognized role in the maintenance of the physiological balance and homeostasis seems to be severely disturbed throughout the carcinogenesis process. Indeed, the modus operandi of cancer implies the highjack of the EV signaling network to support tumor progression in many (if not all) human tumor malignancies. We have reviewed the current evidence for the role of EVs in affecting cancer hallmark traits by: (i) promoting cell proliferation and escape from apoptosis, (ii) sustaining angiogenesis, (iii) contributing to cancer cell invasion and metastasis, (iv) reprogramming energy metabolism, (v) transferring mutations, and (vi) modulating the tumor microenvironment (TME) by evading immune response and promoting inflammation. Special emphasis was given to the role of EVs in the transfer of drug resistant traits and to the EV cargo responsible for this transfer, both between cancer cells or between the microenvironment and tumor cells. Finally, we reviewed evidence for the increased release of EVs by drug resistant cells. A timely and comprehensive understanding of how tumor EVs facilitate tumor initiation, progression, metastasis and drug resistance is instrumental for the development of innovative EV-based therapeutic approaches for cancer.
Collapse
|