1
|
Kim S, Kim YK, Kim S, Choi YS, Lee I, Joo H, Kim J, Kwon M, Park S, Jo MK, Choi Y, D'Souza T, Jung JW, Zakhem E, Lenzini S, Woo J, Choi H, Park J, Park SY, Kim GB, Nam GH, Kim IS. Dual-mode action of scalable, high-quality engineered stem cell-derived SIRPα-extracellular vesicles for treating acute liver failure. Nat Commun 2025; 16:1903. [PMID: 39988725 PMCID: PMC11847939 DOI: 10.1038/s41467-025-57133-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
Acute liver failure (ALF) is a life-threatening condition caused by rapid hepatocyte death and impaired liver regeneration. Here we show that extracellular vesicles engineered to express Signal Regulatory Protein Alpha (SIRP-EVs), produced via a scalable 3D bioreactor process with high yield and purity, exhibit significant therapeutic potential by targeting damaged cells and promoting tissue repair. SIRP-EVs block CD47, a crucial inhibitory signal on necroptotic cells, to enhance macrophage-mediated clearance of dying hepatocytes. They also deliver regenerative cargo from mesenchymal stem cells, reprogramming macrophages to support liver regeneration. In male animal models, SIRP-EVs significantly reduce liver injury markers and improve survival, demonstrating their dual-function therapeutic efficacy. By integrating the resolution of necroptosis with regenerative macrophage reprogramming, SIRP-EVs represent a promising platform for restoring liver function. These findings support the development of EV-based in vivo macrophage reprogramming therapies for ALF and other inflammation-driven diseases, paving the way for the clinical application of engineered EV therapeutics.
Collapse
Grants
- This research was funded by National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (Grant Number: RS-2017-NR022964).
- This research was supported by SHIFTBIO INC., Korean Fund for Regenerative Medicine funded by Ministry of Science and ICT, and Ministry of Health and Welfare (Grant Number: 23C0111L1), a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant Number: RS-2023-KH136648), and a grant of the BIG3 Project, funded by the Ministry of SMEs and Startups, Republic of Korea (Grant Number: RS-2022-TI022422).
- This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant Number: RS-2023-KH136648; RS-2023-KH140007).
Collapse
Affiliation(s)
| | | | | | | | - Inkyu Lee
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Hyemin Joo
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | | | - Minjeong Kwon
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seryoung Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Min Kyoung Jo
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | - Jiwan Woo
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Portrai, Inc, Seoul, Republic of Korea
| | | | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju, Republic of Korea
| | | | - Gi-Hoon Nam
- SHIFTBIO INC, Seoul, Republic of Korea.
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
| |
Collapse
|
2
|
Okuyan HM, Coşkun A, Begen MA. Current status, opportunities, and challenges of exosomes in diagnosis and treatment of osteoarthritis. Life Sci 2025; 362:123365. [PMID: 39761740 DOI: 10.1016/j.lfs.2024.123365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/22/2024] [Accepted: 12/31/2024] [Indexed: 01/19/2025]
Abstract
Osteoarthritis (OA) is a progressive joint disease that is a frequent reason for pain and physical dysfunction in adults, with enormous social and economic burden. Although ongoing scientific efforts in recent years have made considerable progress towards understanding of the disease's molecular mechanism, the pathogenesis of OA is still not fully known, and its clinical challenge remains. Thus, elucidating molecular events underlying the initiation and progression of OA is crucial for developing novel diagnostic and therapeutic approaches that could facilitate effective clinical management of the illness. Exosomes, extracellular vesicles containing various cellular components with approximately a diameter of 100 nm, act as essential mediators in physiological and pathological processes by modulating cell-to-cell communications. Exosomes have crucial roles in biological events such as intercellular communication, regulation of gene expression, apoptosis, inflammation, immunity, maturation and differentiation due to their inner composition, which includes nucleic acids, proteins, and lipids. We focus on the roles of exosomes in OA pathogenesis and discuss how they might be used in clinical practice for OA diagnosis and treatment. Our paper not only provides a comprehensive review of exosomes in OA but also contributes to the development efforts of diagnostic and therapeutic tools for OA.
Collapse
Affiliation(s)
- Hamza Malik Okuyan
- Department of Physiotherapy and Rehabilitation - Faculty of Health Sciences, Biomedical Technologies Application and Research Center, Physiotherapy and Rehabilitation Application and Research Center, Sakarya University of Applied Sciences, Sakarya, Türkiye.
| | - Ayça Coşkun
- Department of Physiotherapy and Rehabilitation - Faculty of Health Sciences, Physiotherapy and Rehabilitation Application and Research Center, Sakarya University of Applied Sciences, Sakarya, Türkiye
| | - Mehmet A Begen
- Department of Epidemiology and Biostatistics-Schulich School of Medicine and Dentistry, Ivey Business School, University of Western Ontario, London, ON, Canada
| |
Collapse
|
3
|
Kareem RA, Sameer HN, Yaseen A, Athab ZH, Adil M, Ahmed HH. A review of the immunomodulatory properties of mesenchymal stem cells and their derived extracellular vesicles in small-cell and non-small-cell lung cancer cells. Int Immunopharmacol 2025; 146:113848. [PMID: 39689606 DOI: 10.1016/j.intimp.2024.113848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024]
Abstract
Among the most challenging diseases to treat is lung cancer (LC). While immunotherapy has a checkered history, it has lately shown great promise in the treatment of LC, and interest in this promising new approach is on the rise around the globe. Immunotherapy using mesenchymal stem cells (MSCs) is gaining popularity. Regenerative medicine, cell therapy, and immune modulation are three areas that have shown significant interest in MSCs. More than that, MSCs have recently attracted attention as potential anti-cancer drug delivery vehicles due to their inherent ability to go home to tumor locations. Making MSCs a double-edged sword in the fight against neoplastic illnesses, they are also known to impart pro-oncogenic properties. Additionally, multiple studies have proposed extracellular vesicles (EVs) secreted by MSCs as a potential therapeutic agent or method for delivering anti-cancer drugs. However, there has been conflicting evidence regarding the impact of MSCs or MSC-EV on the behavior of cancer cells, and the exact mechanism for this effect is still unknown. Our research has focused on MSCs and their key characteristics, such as their immunomodulatory capabilities for cancer therapy. Our research has also explored the potential of MSCs and their derivatives to treat small-cell and non-small-cell lung cancers (NSCLC and SCLC, respectively) by leveraging MSCs' immunomodulatory characteristics. At the end of this article, we covered the pros and cons of this therapy procedure, as well as what researchers want to do in the future to make it more suitable for clinical application in LC treatment.
Collapse
Affiliation(s)
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | | | |
Collapse
|
4
|
Lei R, Ren S, Ye H, Cui Z. Purification of mesenchymal stromal cell-derived small extracellular vesicles using ultrafiltration. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70030. [PMID: 39830832 PMCID: PMC11739894 DOI: 10.1002/jex2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/03/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
Mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) are pivotal for the curative effects of mesenchymal stromal cells, but their translation into clinical products is hindered by the technical challenges of scaled production and purification. Ultrafiltration, a pressure-driven membrane separation method, is well known as an efficient, scalable, and cost-effective approach for bioseparation. However, there has been little study so far that comprehensively evaluates the potential application of ultrafiltration for scaled sEV isolation and purification. In this study, the feasibility and effectiveness of ultrafiltration for MSC-sEV isolation and purification are studied, and the effects of key process design and operational parameters, including the membrane pore size, transmembrane pressure (TMP), stirring speed (shear rate), feed concentration, are quantified using a stirred cell setup. Results revealed that 500 kDa molecular weight cut-off (MWCO) polyethersulfone membrane demonstrated superior suitability for MSC-sEV separation, yielding higher purity and productivity compared to 100 and 300 kDa MWCO membranes of the same material. The MSC-sEV productivity and purity could also be improved by applying a moderate stirring speed and lower operational pressure, respectively. Isovolumetric diafiltration was incorporated to enhance the purity of MSC-sEVs, successfully removing about 99% of protein contaminants by six diafiltration volumes (DVs). Subsequently, a fed-batch ultra-diafiltration (UF/DF) process with optimised filtration parameters was developed and compared with the currently most used ultracentrifugation (UC) method, showing exceptional effectiveness and performance in the isolation of MSC-sEVs: it increased the recovery of MSC-sEV from 20.59% to 60.88% (about three folds increase) and nearly doubled the purity, while also reducing processing time from over 4 h to 3.5 h, with a potential further reduction to less than 2.5 h through automation. The study concludes that ultrafiltration could be a promising method for both lab-scale preparation and industrial-scale manufacture of MSC-sEVs, offering advantages of high recovery, scalability, fast, and cost-effectiveness.
Collapse
Affiliation(s)
- Rui Lei
- Institute of Biomedical Engineering, Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Shuai Ren
- Institute of Biomedical Engineering, Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering ScienceUniversity of OxfordOxfordUK
| |
Collapse
|
5
|
Liu W, Wang X, Chen Y, Yuan J, Zhang H, Jin X, Jiang Y, Cao J, Wang Z, Yang S, Wang B, Wu T, Li J. Distinct molecular properties and functions of small EV subpopulations isolated from human umbilical cord MSCs using tangential flow filtration combined with size exclusion chromatography. J Extracell Vesicles 2025; 14:e70029. [PMID: 39783889 PMCID: PMC11714183 DOI: 10.1002/jev2.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/12/2025] Open
Abstract
As functional derivatives of mesenchymal stem cells (MSCs), small extracellular vesicles (sEVs) have garnered significant attention and application in regenerative medicine. However, the technical limitations for large-scale isolation of sEVs and their heterogeneous nature have added complexity to their applications. It remains unclear if the heterogeneous sEVs represent different aspects of MSCs functions. Here, we provide a method for the large-scale production of sEVs subpopulations derived from human umbilical cord mesenchymal stem cells (HucMSCs), utilizing tangential flow filtration combined with size exclusion chromatography. The resulting subpopulations, S1-sEVs and S2-sEVs, exhibited stable variations in size, membrane-marked proteins, and carrying cargos, thereby displaying distinct functions both in vitro and in animal disease models. S1-sEVs, that highly expressed CD9, HRS and GPC1, demonstrated a greater immunomodulatory impact, while S2-sEVs with enriched expression of CD63 and FLOT1/2 possessed enhanced capacities in promoting cell proliferation and angiogenesis. These discrepancies are attributed to the specific proteins and miRNAs they contain. Further investigation revealed that the two distinct sEVs subpopulations corresponded to different biological processes: the ESCRT pathway (S1-sEVs) and the ESCRT-independent pathway represented by lipid rafts (S2-sEVs). Therefore, we propose the potential for large-scale isolation and purification of sEVs subpopulations from HucMSCs with distinct functions. This approach may provide advantages for targeted therapeutic interventions in various MSC indications.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xinyu Wang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Yating Chen
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Jiapei Yuan
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Huiyu Zhang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Xin Jin
- Wuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiChina
| | - Yuying Jiang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Junjing Cao
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
| | - Zibin Wang
- Center for Analysis and TestingNanjing Medical UniversityNanjingChina
| | - Shuo Yang
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
- Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical CenterNanjing Medical UniversityNanjingChina
| | - Bingwei Wang
- Department of PharmacologyNanjing University of Chinese MedicineNanjingChina
| | - Tinghe Wu
- Kornelis Bio‐pharmaceutical Company LimitedNanjingChina
| | - Jing Li
- State Key Laboratory of Reproductive Medicine and offspring healthNanjing Medical UniversityNanjingChina
- Innovation Center of Suzhou Nanjing Medical UniversitySuzhouChina
| |
Collapse
|
6
|
Lu X, Wang Y, Piao C, Li P, Cao L, Liu T, Ma Y, Wang H. Exosomes Derived from Adipose Mesenhymal Stem Cells Ameliorate Lipid Metabolism Disturbances Following Liver Ischemia-Reperfusion Injury in Miniature Swine. Int J Mol Sci 2024; 25:13069. [PMID: 39684778 DOI: 10.3390/ijms252313069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
The liver plays a crucial role in regulating lipid metabolism. Our study examined the impact of Exosomes derived from adipose mesenchymal stem cells (ADSCs-Exo) on lipid metabolism following liver ischemia-reperfusion injury (IRI) combined with partial hepatectomy. We developed a miniature swine model for a minimally invasive hemi-hepatectomy combined with liver IRI. In this study, we administered PBS, ADSCs-Exo, and adipose-derived stem cells (ADSCs) individually through the portal vein. Before and after surgery, we evaluated various factors including hepatocyte ultrastructure, lipid accumulation in liver tissue, and expression levels of genes and proteins associated with lipid metabolism. In addition, we measured serum and liver tissue levels of high-density lipoprotein (HDL), low-density lipoprotein (LDL), triglycerides (TG), and total cholesterol (CHOL). TEM and oil red O stain indicated a significant reduction in liver steatosis following ADSCs-Exo treatment, which also elevated serum levels of HDL, LDL, TG, and CHOL. Additionally, ADSCs-Exo have been shown to significantly decrease serum concentrations of HDL, LDL, TG, and CHOL in the liver (p < 0.05). Finally, ADSCs-Exo significantly downregulated lipid synthesis-related genes and proteins, including SREBP-1, SREBP-2, ACC1, and FASN (p < 0.05), while upregulating lipid catabolism-related genes and proteins, such as PPAR-α and ACOX1 (p < 0.05). ADSCs-Exo as a cell-free therapy highlights its therapeutic potential in hepatic lipid metabolism abnormalities.
Collapse
Affiliation(s)
- Xiangyu Lu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Yue Wang
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
| | - Chenxi Piao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Pujun Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Lei Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Harbin 150030, China
| |
Collapse
|
7
|
Wa Q, Luo Y, Tang Y, Song J, Zhang P, Linghu X, Lin S, Li G, Wang Y, Wen Z, Huang S, Xu W. Mesoporous bioactive glass-enhanced MSC-derived exosomes promote bone regeneration and immunomodulation in vitro and in vivo. J Orthop Translat 2024; 49:264-282. [PMID: 39524151 PMCID: PMC11550139 DOI: 10.1016/j.jot.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Background Exosomes produced by mesenchymal stem cells (MSCs) have vascular generative properties and are considered new effective candidates for the treatment of bone defects as alternatives to cell therapy. Improving the pro-regenerative function and efficacy of exosomes has been a popular research topic in the field of orthopaedics. Methods We prepared mesoporous bioactive glass (mBG) microspheres via the template method. The ionic products of mBGs used to treat MSCs were extracted, and the effects of exosomes secreted by MSCs on osteoblast (OB) and macrophage (MP) behaviour and bone defect repair were observed in vivo (Micro-CT, H&E, Masson, and immunofluorescence staining for BMP2, COL1, VEGF, CD31, CD163, and iNOS). Results The mBG spheres were successfully prepared, and the Exo-mBG were isolated and extracted. Compared with those in the blank and Exo-Con groups, the proliferation and osteogenic differentiation of OBs in the Exo-mBG group were significantly greater. For example, on Day 7, OPN gene expression in the Ctrl-Exo group was 3.97 and 2.83 times greater than that in the blank and Exo-mBG groups, respectively. In a cranial defect rat model, Exo-mBG promoted bone tissue healing and angiogenesis, increased M2 macrophage polarisation and inhibited M1 macrophage polarisation, as verified by micro-CT, H&E staining, Masson staining and immunofluorescence staining. These effects may be due to the combination of a higher silicon concentration and a higher calcium-to-phosphorus ratio in the mBG ionic products. Conclusion This study provides insights for the application of exosomes in cell-free therapy and a new scientific basis and technical approach for the utilisation of MSC-derived exosomes in bone defect repair. The translational potential of this article Our study demonstrated that exosomes produced by mBG-stimulated MSCs have excellent in vitro and in vivo bone-enabling and immunomodulatory functions and provides insights into the use of exosomes in clinical cell-free therapies.
Collapse
Affiliation(s)
- Qingde Wa
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China. Intersection of Xinlong Avenue and Xinpu Avenue, Honghuagang District, Zunyi, Guizhou, 563000, China
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, No.10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
| | - Yongxiang Luo
- Marshall Biomedical Engineering Laboratory, Shenzhen University, No. 3688 Nanhai Avenue, Nanshan District, Shenzhen, Guangdong, 518060, China
| | - Yubo Tang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan Second Road, Guangzhou, Guangdong, 510080, China
| | - Jiaxiang Song
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China. Intersection of Xinlong Avenue and Xinpu Avenue, Honghuagang District, Zunyi, Guizhou, 563000, China
| | - Penghui Zhang
- Department of Orthopaedics, Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Xitao Linghu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China. Intersection of Xinlong Avenue and Xinpu Avenue, Honghuagang District, Zunyi, Guizhou, 563000, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yixiao Wang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China, No. 98 Fenghuang North Road, Huichuan District, Zunyi City, Guizhou, 563002, China
| | - Zhenyu Wen
- Zunyi Medical University, No. 1 Campus, Xinpu New District, Zunyi City, Guizhou, 563000, China
| | - Shuai Huang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong, 510260, China
| | - Weikang Xu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, No.10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307 Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong, 510500, China
- Guangdong Chinese Medicine Intelligent Diagnosis and Treatment Engineering Technology Research Center, No. 10 Shiliugang Road, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong, 510316, China
| |
Collapse
|
8
|
Louro AF, Meliciano A, Alves PM, Costa MHG, Serra M. A roadmap towards manufacturing extracellular vesicles for cardiac repair. Trends Biotechnol 2024; 42:1305-1322. [PMID: 38653588 DOI: 10.1016/j.tibtech.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
For the past two decades researchers have linked extracellular vesicle (EV)-mediated mechanisms to various physiological and pathological processes in the heart, such as immune response regulation, fibrosis, angiogenesis, and the survival and growth of cardiomyocytes. Although use of EVs has gathered momentum in the cardiac field, several obstacles in both upstream and downstream processes during EV manufacture need to be addressed before clinical success can be achieved. Low EV yields obtained in small-scale cultures deter clinical translation, as mass production is a prerequisite to meet therapeutic doses. Moreover, standardizing EV manufacture is critical given the inherent heterogeneity of EVs and the constraints of current isolation techniques. In this review, we discuss the critical steps for the large-scale manufacturing of high-potency EVs for cardiac therapies.
Collapse
Affiliation(s)
- Ana F Louro
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana Meliciano
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Marta H G Costa
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
9
|
Matloob A, Gu X, Rehman Sheikh A, Javed M, Fang Z, Luo Z. Plant exosomes‐like nano‐vesicles: Characterization, functional food potential, and emerging therapeutic applications as a nano medicine. FOOD SAFETY AND HEALTH 2024; 2:429-450. [DOI: 10.1002/fsh3.12060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 01/05/2025]
Abstract
AbstractPlant cells release exosome‐like nanovesicles (PENVs), which are small, membrane‐bound vesicles secreted by cells for intercellular interactions. These vesicles, rich in biologically active substances, are crucial for information transmission, intercellular interaction, and organism homeostasis conservation. They can also be used for treating diseases as large‐scale drug carriers due to their vesicular architecture. This study explores the isolation, potential of nanovesicles in creating bio‐therapeutic and drug‐delivery nano‐platforms to address clinical challenges. The bio‐therapeutic roles of PENVs, include immunomodulation, antitumor, regenerative impacts, wound healing, anti‐fibrosis, whitening effects, and treatment of intestinal flora disorders. This study also deliberates the potential for designing these nanovesicles into effective, safe, and non‐immunogenic nano‐vectors to carry drugs. PENVs may offer a cutting‐edge platform for the creation of functional foods and nutraceuticals. They might be employed to encapsulate certain bioactive substances produced from plants, offering tailored health privileges. It also elucidates the potential for the development of therapeutic and provision nano‐platforms based on PENVs in clinical applications.
Collapse
Affiliation(s)
- Anam Matloob
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Xinya Gu
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Arooj Rehman Sheikh
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Miral Javed
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Zhongxiang Fang
- School of Agriculture, Food and Ecosystem Sciences Faculty of Science The University of Melbourne Melbourne Victoria Australia
| | - Zisheng Luo
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
- Key Laboratory of Ago‐Products Postharvest Handing of Ministry of Agriculture and Rural Affairs Hangzhou China
| |
Collapse
|
10
|
Kim HK, Choi Y, Kim KH, Byun Y, Kim TH, Kim JH, An SH, Bae D, Choi MK, Lee M, Kang G, Chung J, Kim S, Kwon K. Scalable production of siRNA-encapsulated extracellular vesicles for the inhibition of KRAS-mutant cancer using acoustic shock waves. J Extracell Vesicles 2024; 13:e12508. [PMID: 39323378 PMCID: PMC11424982 DOI: 10.1002/jev2.12508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 07/16/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as a potential delivery vehicle for nucleic-acid-based therapeutics, but challenges related to their large-scale production and cargo-loading efficiency have limited their therapeutic potential. To address these issues, we developed a novel "shock wave extracellular vesicles engineering technology" (SWEET) as a non-genetic, scalable manufacturing strategy that uses shock waves (SWs) to encapsulate siRNAs in EVs. Here, we describe the use of the SWEET platform to load large quantities of KRASG12C-targeting siRNA into small bovine-milk-derived EVs (sBMEVs), with high efficiency. The siRNA-loaded sBMEVs effectively silenced oncogenic KRASG12C expression in cancer cells; they inhibited tumour growth when administered intravenously in a non-small cell lung cancer xenograft mouse model. Our study demonstrates the potential for the SWEET platform to serve as a novel method that allows large-scale production of cargo-loaded EVs for use in a wide range of therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - DaeHo Bae
- Exollence Co., Ltd.SeoulRepublic of Korea
| | | | | | - Gwansuk Kang
- Division of Gastroenterology and Hepatology, School of MedicineStanford UniversityStanfordCaliforniaUSA
| | | | | | - Kihwan Kwon
- Exollence Co., Ltd.SeoulRepublic of Korea
- Department of Internal Medicine, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| |
Collapse
|
11
|
Salerno S, Piscioneri A, Morelli S, Gori A, Provasi E, Gagni P, Barile L, Cretich M, Chiari M, De Bartolo L. Extracellular vesicles selective capture by peptide-functionalized hollow fiber membranes. J Colloid Interface Sci 2024; 667:338-349. [PMID: 38640653 DOI: 10.1016/j.jcis.2024.04.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024]
Abstract
Recently, membrane devices and processes have been applied for the separation and concentration of subcellular components such as extracellular vesicles (EVs), which play a diagnostic and therapeutic role in many pathological conditions. However, the separation and isolation of specific EV populations from other components found in biological fluids is still challenging. Here, we developed a peptide-functionalized hollow fiber (HF) membrane module to achieve the separation and enrichment of highly pure EVs derived from the culture media of human cardiac progenitor cells. The strategy is based on the functionalization of PSf HF membrane module with BPt, a peptide sequence able to bind nanovesicles characterized by highly curved membranes. HF membranes were modified by a nanometric coating with a copoly azide polymer to limit non-specific interactions and to enable the conjugation with peptide ligand by click chemistry reaction. The BPt-functionalized module was integrated into a TFF process to facilitate the design, rationalization, and optimization of EV isolation. This integration combined size-based transport of species with specific membrane sensing ligands. The TFF integrated BPt-functionalized membrane module demonstrated the ability to selectively capture EVs with diameter < 200 nm into the lumen of fibers while effectively removing contaminants such as albumin. The captured and released EVs contain the common markers including CD63, CD81, CD9 and syntenin-1. Moreover, they maintained a round shape morphology and structural integrity highlighting that this approach enables EVs concentration and purification with low shear stress. Additionally, it achieved the removal of contaminants such as albumin with high reliability and reproducibility, reaching a removal of 93%.
Collapse
Affiliation(s)
- Simona Salerno
- Institute on Membrane Technology, National Research Council of Italy, ITM-CNR, via P. Bucci, cubo 17/C, I-87036 Rende (CS), Italy
| | - Antonella Piscioneri
- Institute on Membrane Technology, National Research Council of Italy, ITM-CNR, via P. Bucci, cubo 17/C, I-87036 Rende (CS), Italy
| | - Sabrina Morelli
- Institute on Membrane Technology, National Research Council of Italy, ITM-CNR, via P. Bucci, cubo 17/C, I-87036 Rende (CS), Italy
| | - Alessandro Gori
- Institute of Chemical Sciences and Technologies "G. Natta", National Research Council of Italy, SCITEC-CNR, Via Mario Bianco 9, 20131, Milan, Italy
| | - Elena Provasi
- Lugano Cell Factory, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, via Tesserete 48, 6900 Lugano, Switzerland
| | - Paola Gagni
- Institute of Chemical Sciences and Technologies "G. Natta", National Research Council of Italy, SCITEC-CNR, Via Mario Bianco 9, 20131, Milan, Italy
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa 5, 6500 Bellinzona, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Via Buffi 13, 6900 Lugano, Switzerland
| | - Marina Cretich
- Institute of Chemical Sciences and Technologies "G. Natta", National Research Council of Italy, SCITEC-CNR, Via Mario Bianco 9, 20131, Milan, Italy
| | - Marcella Chiari
- Institute of Chemical Sciences and Technologies "G. Natta", National Research Council of Italy, SCITEC-CNR, Via Mario Bianco 9, 20131, Milan, Italy
| | - Loredana De Bartolo
- Institute on Membrane Technology, National Research Council of Italy, ITM-CNR, via P. Bucci, cubo 17/C, I-87036 Rende (CS), Italy.
| |
Collapse
|
12
|
Zhang Z, Wu W, Li M, Du L, Li J, Yin X, Zhang W. Mesenchymal stem cell–derived extracellular vesicles: A novel nanoimmunoregulatory tool in musculoskeletal diseases. NANO TODAY 2024; 57:102343. [DOI: 10.1016/j.nantod.2024.102343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Saari H, Marttila H, Poranen MM, Oksanen HM, Zini J, Laitinen S. Inline Raman Spectroscopy Provides Versatile Molecular Monitoring for Platelet Extracellular Vesicle Purification with Anion-Exchange Chromatography. Int J Mol Sci 2024; 25:8130. [PMID: 39125704 PMCID: PMC11311901 DOI: 10.3390/ijms25158130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Extracellular vesicles (EVs) are relatively recently discovered biological nanoparticles that mediate intercellular communication. The development of new methods for the isolation and characterization of EVs is crucial to support further studies on these small and structurally heterogenous vesicles. New scalable production methods are also needed to meet the needs of future therapeutic applications. A reliable inline detection method for the EV manufacturing process is needed to ensure reproducibility and to identify any possible variations in real time. Here, we demonstrate the use of an inline Raman detector in conjunction with anion exchange chromatography for the isolation of EVs from human platelets. Anion-exchange chromatography can be easily coupled with multiple inline detectors and provides an alternative to size-based methods for separating EVs from similar-sized impurities, such as lipoprotein particles. Raman spectroscopy enabled us to identify functional groups in EV samples and trace EVs and impurities in different stages of the process. Our results show a notable separation of impurities from the EVs during anion-exchange chromatography and demonstrate the power of inline Raman spectroscopy. Compared to conventional EV analysis methods, the inline Raman approach does not require hands-on work and can provide detailed, real-time information about the sample and the purification process.
Collapse
Affiliation(s)
- Heikki Saari
- Finnish Red Cross, Blood Service, Härkälenkki 13, 01730 Vantaa, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, 00790 Helsinki, Finland
| | - Heli Marttila
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9, 00790 Helsinki, Finland
| | - Minna M. Poranen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9, 00790 Helsinki, Finland
| | - Hanna M. Oksanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9, 00790 Helsinki, Finland
| | - Jacopo Zini
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, 00790 Helsinki, Finland
- Timegate Instruments Ltd., Tutkijantie 7, 90590 Oulu, Finland
| | - Saara Laitinen
- Finnish Red Cross, Blood Service, Härkälenkki 13, 01730 Vantaa, Finland
| |
Collapse
|
14
|
Bae J, Lee C, Jung D, Yea K, Song B, Lee H, Baek M. Extracellular vesicle isolation and counting system (EVics) based on simultaneous tandem tangential flow filtration and large field-of-view light scattering. J Extracell Vesicles 2024; 13:e12479. [PMID: 38978321 PMCID: PMC11231039 DOI: 10.1002/jev2.12479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024] Open
Abstract
Although the isolation and counting of small extracellular vesicles (sEVs) are essential steps in sEV research, an integrated method with scalability and efficiency has not been developed. Here, we present a scalable and ready-to-use extracellular vesicle (EV) isolation and counting system (EVics) that simultaneously allows isolation and counting in one system. This novel system consists of (i) EVi, a simultaneous tandem tangential flow filtration (TFF)-based EV isolation component by applying two different pore-size TFF filters, and (ii) EVc, an EV counting component using light scattering that captures a large field-of-view (FOV). EVi efficiently isolated 50-200 nm-size sEVs from 15 µL to 2 L samples, outperforming the current state-of-the-art devices in purity and speed. EVc with a large FOV efficiently counted isolated sEVs. EVics enabled early observations of sEV secretion in various cell lines and reduced the cost of evaluating the inhibitory effect of sEV inhibitors by 20-fold. Using EVics, sEVs concentrations and sEV PD-L1 were monitored in a 23-day cancer mouse model, and 160 clinical samples were prepared and successfully applied to diagnosis. These results demonstrate that EVics could become an innovative system for novel findings in basic and applied studies in sEV research.
Collapse
Affiliation(s)
- Ju‐Hyun Bae
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC)School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Chan‐Hyeong Lee
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC)School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Dokyung Jung
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC)School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Kyungmoo Yea
- Department of New BiologyDGISTDaeguRepublic of Korea
- New Biology Research CenterDGISTDaeguRepublic of Korea
| | - Byoung‐Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and BiophysicsNational Institute on Alcohol Abuse and Alcoholism, NIHBethesdaMarylandUSA
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of Radiology, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Moon‐Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC)School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| |
Collapse
|
15
|
Zhou W, Wang X, Dong Y, Gao P, Zhao X, Wang M, Wu X, Shen J, Zhang X, Lu Z, An W. Stem cell-derived extracellular vesicles in the therapeutic intervention of Alzheimer's Disease, Parkinson's Disease, and stroke. Theranostics 2024; 14:3358-3384. [PMID: 38855176 PMCID: PMC11155406 DOI: 10.7150/thno.95953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024] Open
Abstract
With the increase in the aging population, the occurrence of neurological disorders is rising. Recently, stem cell therapy has garnered attention due to its convenient sourcing, minimal invasiveness, and capacity for directed differentiation. However, there are some disadvantages, such as poor quality control, safety assessments, and ethical and logistical issues. Consequently, scientists have started to shift their attention from stem cells to extracellular vesicles due to their similar structures and properties. Beyond these parallels, extracellular vesicles can enhance biocompatibility, facilitate easy traversal of barriers, and minimize side effects. Furthermore, stem cell-derived extracellular vesicles can be engineered to load drugs and modify surfaces to enhance treatment outcomes. In this review, we summarize the functions of native stem cell-derived extracellular vesicles, subsequently review the strategies for the engineering of stem cell-derived extracellular vesicles and their applications in Alzheimer's disease, Parkinson's disease, and stroke, and discuss the challenges and solutions associated with the clinical translation of stem cell-derived extracellular vesicles.
Collapse
Affiliation(s)
- Wantong Zhou
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xudong Wang
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Yumeng Dong
- Capital Medical University, 10 Xitoutiao, Youanmenwai Street, Beijing 100069, China
| | - Peifen Gao
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xian Zhao
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Mengxia Wang
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xue Wu
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Jiuheng Shen
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wenlin An
- National Vaccine Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| |
Collapse
|
16
|
Menjivar NG, Oropallo J, Gebremedhn S, Souza LA, Gad A, Puttlitz CM, Tesfaye D. MicroRNA Nano-Shuttles: Engineering Extracellular Vesicles as a Cutting-Edge Biotechnology Platform for Clinical Use in Therapeutics. Biol Proced Online 2024; 26:14. [PMID: 38773366 PMCID: PMC11106895 DOI: 10.1186/s12575-024-00241-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, membranous transporters of various active biomolecules with inflicting phenotypic capabilities, that are naturally secreted by almost all cells with a promising vantage point as a potential leading drug delivery platform. The intrinsic characteristics of their low toxicity, superior structural stability, and cargo loading capacity continue to fuel a multitude of research avenues dedicated to loading EVs with therapeutic and diagnostic cargos (pharmaceutical compounds, nucleic acids, proteins, and nanomaterials) in attempts to generate superior natural nanoscale delivery systems for clinical application in therapeutics. In addition to their well-known role in intercellular communication, EVs harbor microRNAs (miRNAs), which can alter the translational potential of receiving cells and thus act as important mediators in numerous biological and pathological processes. To leverage this potential, EVs can be structurally engineered to shuttle therapeutic miRNAs to diseased recipient cells as a potential targeted 'treatment' or 'therapy'. Herein, this review focuses on the therapeutic potential of EV-coupled miRNAs; summarizing the biogenesis, contents, and function of EVs, as well as providing both a comprehensive discussion of current EV loading techniques and an update on miRNA-engineered EVs as a next-generation platform piloting benchtop studies to propel potential clinical translation on the forefront of nanomedicine.
Collapse
Affiliation(s)
- Nico G Menjivar
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jaiden Oropallo
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
- Orthopaedic Research Center (ORC), Translational Medicine Institute (TMI), Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Samuel Gebremedhn
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- J.R. Simplot Company, 1099 W. Front St, Boise, ID, 83702, USA
| | - Luca A Souza
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, 225 Av. Duque de Caxias Norte, Pirassununga, SP, 13635-900, Brazil
| | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, 12613, Egypt
| | - Christian M Puttlitz
- Orthopaedic Bioengineering Research Laboratory (OBRL), Translational Medicine Institute (TMI), Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
17
|
Iannotta D, A A, Lai A, Nair S, Koifman N, Lappas M, Salomon C, Wolfram J. Chemically-Induced Lipoprotein Breakdown for Improved Extracellular Vesicle Purification. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307240. [PMID: 38100284 DOI: 10.1002/smll.202307240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Extracellular vesicles (EVs) are nanosized biomolecular packages involved in intercellular communication. EVs are released by all cells, making them broadly applicable as therapeutic, diagnostic, and mechanistic components in (patho)physiology. Sample purity is critical for correctly attributing observed effects to EVs and for maximizing therapeutic and diagnostic performance. Lipoprotein contaminants represent a major challenge for sample purity. Lipoproteins are approximately six orders of magnitude more abundant in the blood circulation and overlap in size, shape, and density with EVs. This study represents the first example of an EV purification method based on the chemically-induced breakdown of lipoproteins. Specifically, a styrene-maleic acid (SMA) copolymer is used to selectively breakdown lipoproteins, enabling subsequent size-based separation of the breakdown products from plasma EVs. The use of the polymer followed by tangential flow filtration or size-exclusion chromatography results in improved EV yield, preservation of EV morphology, increased EV markers, and reduced contaminant markers. SMA-based EV purification enables improved fluorescent labeling, reduces interactions with macrophages, and enhances accuracy, sensitivity, and specificity to detect EV biomarkers, indicating benefits for various downstream applications. In conclusion, SMA is a simple and effective method to improve the purity and yield of plasma-derived EVs, which favorably impacts downstream applications.
Collapse
Affiliation(s)
- Dalila Iannotta
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Amruta A
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Na'ama Koifman
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Martha Lappas
- University of Melbourne, Department of Obstetrics and Gynaecology, Australia, and Mercy Hospital for Women, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Joy Wolfram
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| |
Collapse
|
18
|
Dai C, Xu Q, Li L, Liu Y, Qu S. Milk Extracellular Vesicles: Natural Nanoparticles for Enhancing Oral Drug Delivery against Bacterial Infections. ACS Biomater Sci Eng 2024; 10:1988-2000. [PMID: 38529792 DOI: 10.1021/acsbiomaterials.3c01824] [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] [Indexed: 03/27/2024]
Abstract
Oral drug delivery is typically preferred as a therapeutic intervention due to the complexities and expenses associated with intravenous administration. However, some drugs are poorly absorbed orally, requiring intravenous administration to bypass the gastrointestinal tract and deliver the drug directly into the bloodstream. Thus, there is an urgent need to develop novel drug delivery platforms to overcome the challenges of oral drug delivery with low solubility, low permeability, oral degradation, and low bioavailability. Advances in extracellular vesicles (EVs) as natural carriers have provided emerging approaches to improve potential therapeutic applications. Milk not only contains traditional nutrients but is also rich in EVs. In this Review, we focus mainly on the purification of milk EVs (mEVs), their safety, and the advantages of mEV-based drug carriers in combatting intestinal infections. Additionally, we summarize several advantages of mEVs over conventional synthetic carriers, such as low immunogenicity, high biocompatibility, and the ability to transfer bioactive molecules between cells. Considering the unmet gaps of mEVs in clinical translation, it is essential to review the cargo loading into mEVs and future perspectives for their use as natural drug carriers for oral delivery. This overview of mEV-based drug carriers for oral delivery sheds light on alternative approaches to treat clinical infections associated with intestinal pathogens and the development of novel oral delivery systems.
Collapse
Affiliation(s)
- Cunchun Dai
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Qingjun Xu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ying Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
19
|
Leung J, Pollalis D, Nair GKG, Bailey JK, Pennington BO, Khan AI, Kelly KR, Yeh AK, Sundaram KS, Clegg DO, Peng CC, Xu L, Lee SY. Isolation and Characterization of Extracellular Vesicles Through Orthogonal Approaches for the Development of Intraocular EV Therapy. Invest Ophthalmol Vis Sci 2024; 65:6. [PMID: 38466285 PMCID: PMC10929743 DOI: 10.1167/iovs.65.3.6] [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: 12/01/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Purpose Isolating extracellular vesicles (EVs) with high yield, replicable purity, and characterization remains a bottleneck in the development of EV therapeutics. To address these challenges, the current study aims to establish the necessary framework for preclinical and clinical studies in the development of stem cell-derived intraocular EV therapeutics. Methods Small EVs (sEVs) were separated from the conditioned cell culture medium (CCM) of the human embryogenic stem cell-derived fully polarized retinal pigment epithelium (hESC-RPE-sEV) by a commercially available microfluidic tangential flow filtration (TFF) device ExoDisc (ED) or differential ultracentrifugation (dUC). The scaling and concentration capabilities and purity of recovered sEVs were assessed. Size, number, and surface markers of sEVs were determined by orthogonal approaches using multiple devices. Results ED yielded higher numbers of sEVs, ranging from three to eight times higher depending on the measurement device, compared to dUC using the same 5 mL of CCM input. Within the same setting, the purity of ED-recovered hESC-RPE-sEVs was higher than that for dUC-recovered sEVs. ED yielded a higher concentration of particles, which is strongly correlated with the input volume, up to 10 mL (r = 0.98, P = 0.016). Meanwhile, comprehensive characterization profiles of EV surface markers between ED- and dUC-recovered hESC-RPE-sEVs were compatible. Conclusions Our study supports TFF as a valuable strategy for separating sEVs for the development of intraocular EV therapeutics. However, there is a growing need for diverse devices to optimize TFF for use in EV preparation. Using orthogonal approaches in EV characterization remains ideal for reliably characterizing heterogeneous EV.
Collapse
Affiliation(s)
- Justin Leung
- USC Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
| | - Dimitrios Pollalis
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Gopa K. G. Nair
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Jeffrey K. Bailey
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Britney O. Pennington
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Amir I. Khan
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Kaitlin R. Kelly
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Ashley K. Yeh
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- College of Creative Studies, Biology, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Kartik S. Sundaram
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Dennis O. Clegg
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, California, United States
| | - Chen-Ching Peng
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Children's Hospital Los Angeles Vision Center, Los Angeles, California, United States
| | - Liya Xu
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Children's Hospital Los Angeles Vision Center, Los Angeles, California, United States
| | - Sun Young Lee
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, California, United States
- Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| |
Collapse
|
20
|
Zimmerman AJ, de Oliveira GP, Su X, Wood J, Fu Z, Pinckney B, Tigges J, Ghiran I, Ivanov AR. Multimode chromatography-based techniques for high purity isolation of extracellular vesicles from human blood plasma. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e147. [PMID: 38751711 PMCID: PMC11080799 DOI: 10.1002/jex2.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 03/08/2024] [Indexed: 05/18/2024]
Abstract
Extracellular vesicles (EVs) play a pivotal role in various biological pathways, such as immune responses and the progression of diseases, including cancer. However, it is challenging to isolate EVs at high purity from blood plasma and other biofluids due to their low abundance compared to more predominant biomolecular species such as lipoprotein particles and free protein complexes. Ultracentrifugation-based EV isolation, the current gold standard technique, cannot overcome this challenge due to the similar biophysical characteristics of such species. We developed several novel approaches to enrich EVs from plasma while depleting contaminating molecular species using multimode chromatography-based strategies. On average, we identified 716 ± 68 and 1054 ± 35 protein groups in EV isolates from 100 µL of plasma using multimode chromatography- and ultracentrifugation-based techniques, respectively. The developed methods resulted in similar EV isolates purity, providing significant advantages in simplicity, throughput, scalability, and applicability for various downstream analytical and potential clinical applications.
Collapse
Affiliation(s)
- Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Getulio Pereira de Oliveira
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Xianyi Su
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Jacqueline Wood
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Zhengxin Fu
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Brandy Pinckney
- Nano Flow Core Facility, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - John Tigges
- Nano Flow Core Facility, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Ionita Ghiran
- Department of Anesthesia, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| |
Collapse
|
21
|
Roszkowski S. Therapeutic potential of mesenchymal stem cell-derived exosomes for regenerative medicine applications. Clin Exp Med 2024; 24:46. [PMID: 38427086 PMCID: PMC10907468 DOI: 10.1007/s10238-023-01282-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/08/2023] [Indexed: 03/02/2024]
Abstract
Mesenchymal stem cell-derived exosomes have emerged as a promising cell-free therapy for tissue engineering. Compared to intact stem cells, exosomes have advantages like low immunogenicity and ability to carry regenerative cargo. This review examined the potential of exosomes to treat defects in skin, bone and cartilage. In preclinical models, exosomes improved wound healing, stimulated bone regeneration, and enabled cartilage repair by transferring proteins, mRNAs and microRNAs. Their effects were elicited by modulating inflammation, angiogenesis, cell proliferation and matrix synthesis. Exosomes represent a promising cell-free therapy for tissue engineering. However, challenges remain regarding scalable isolation, elucidating mechanisms, and translating this approach to human trials. Understanding these challenges will enable the successful clinical translation of exosomes for regenerative medicine applications.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Debowa St. 3, 85-626, Bydgoszcz, Poland.
| |
Collapse
|
22
|
Khoushab S, Aghmiuni MH, Esfandiari N, Sarvandani MRR, Rashidi M, Taheriazam A, Entezari M, Hashemi M. Unlocking the potential of exosomes in cancer research: A paradigm shift in diagnosis, treatment, and prevention. Pathol Res Pract 2024; 255:155214. [PMID: 38430814 DOI: 10.1016/j.prp.2024.155214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
Exosomes, which are tiny particles released by cells, have the ability to transport various molecules, including proteins, lipids, and genetic material containing non-coding RNAs (ncRNAs). They are associated with processes like cancer metastasis, immunity, and tissue repair. Clinical trials have shown exosomes to be effective in treating cancer, inflammation, and chronic diseases. Mesenchymal stem cells (MSCs) and dendritic cells (DCs) are common sources of exosome production. Exosomes have therapeutic potential due to their ability to deliver cargo, modulate the immune system, and promote tissue regeneration. Bioengineered exosomes could revolutionize disease treatment. However, more research is needed to understand exosomes in tumor growth and develop new therapies. This paper provides an overview of exosome research, focusing on cancer and exosome-based therapies including chemotherapy, radiotherapy, and vaccines. It explores exosomes as a drug delivery system for cancer therapy, highlighting their advantages. The article discusses using exosomes for various therapeutic agents, including drugs, antigens, and RNAs. It also examines challenges with engineered exosomes. Analyzing exosomes for clinical purposes faces limitations in sensitivity, specificity, and purification. On the other hand, Nanotechnology offers solutions to overcome these challenges and unlock exosome potential in healthcare. Overall, the article emphasizes the potential of exosomes for personalized and targeted cancer therapy, while acknowledging the need for further research.
Collapse
Affiliation(s)
- Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Hobabi Aghmiuni
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esfandiari
- Department of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
23
|
Miron RJ, Zhang Y. Understanding exosomes: Part 1-Characterization, quantification and isolation techniques. Periodontol 2000 2024; 94:231-256. [PMID: 37740431 DOI: 10.1111/prd.12520] [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: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 09/24/2023]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with a diameter in the range of 30-150 nm. Their use has gained great momentum recently due to their ability to be utilized as diagnostic tools with a vast array of therapeutic applications. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be investigated. This review article first focuses on understanding exosomes, including their cellular origin, biogenesis, function, and characterization. Thereafter, overviews of the quantification methods and isolation techniques are given with discussion over their potential use as novel therapeutics in regenerative medicine.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
24
|
Chiu YS, Wu KJ, Yu SJ, Wu KL, Hsieh CY, Chou YS, Chen KY, Wang YS, Bae EK, Hung TW, Lin SH, Lin CH, Hsu SC, Wang Y, Chen YH. Transplantation of Exosomes Derived From Human Wharton's Jelly Mesenchymal Stromal Cells Enhances Functional Improvement in Stroke Rats. Cell Transplant 2024; 33:9636897241296366. [PMID: 39624898 PMCID: PMC11613244 DOI: 10.1177/09636897241296366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/28/2024] [Accepted: 10/15/2024] [Indexed: 12/06/2024] Open
Abstract
Cerebral ischemic stroke is a major cerebrovascular disease and the leading cause of adult disability. We and others previously demonstrated that transplantation of human Wharton's jelly mesenchymal stromal cells (WJ-MSCs) attenuated neuronal damage and promoted functional improvement in stroke animals. This study aimed to investigate the protective effects of human WJ-MSC exosome (Exo) transplant in cellular and rat models of cerebral stroke. Administration of Exo significantly antagonized glutamate-mediated neuronal loss and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-X nick end labeling (TUNEL) in rat primary cortical neuronal cultures. Adult male rats underwent a 60-min middle cerebral artery occlusion (MCAo); Exo or vehicle was injected through the tail vein 5-10 min after the MCAo. Two days later, the rats underwent a series of behavioral tests. Stroke rats receiving Exo developed a significant improvement in locomotor function and forelimb strength while reductions in body asymmetry and Bederson's neurological score. After the behavioral test, brain tissues were harvested for histological and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) analyses. Animals receiving Exo had less infarction volume, measured by 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Transplantation of Exo increased the expression of protective neurotrophic factors (BMP7, GDNF) and anti-apoptotic factors (Bcl2, Bcl-xL) in the ischemic brain. These findings suggest that early post-treatment with WJ-MSC Exo, given non-invasively through the vein, improved functional recovery and reduced brain damage in the stroke brain.
Collapse
Affiliation(s)
- Yu-Sung Chiu
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
| | - Kuo-Jen Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kun-Lieh Wu
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
- Department of Electrical Engineering, I-Shou University, Kaohsiung, Taiwan
| | | | | | - Kuan-Yu Chen
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Tsai-Wei Hung
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Hsun Lin
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Hsueh Lin
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Shu-Ching Hsu
- Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
- Immunology Research and Development Center, China Medical University, Taichung City, Taiwan
- Department of Life Sciences, Tzu Chi University, Hualien, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yun-Hsiang Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
25
|
Zhang F, Zhang L, Yu H. Potential Druggability of Mesenchymal Stem/Stromal Cell-derived Exosomes. Curr Stem Cell Res Ther 2024; 19:1195-1209. [PMID: 38523514 DOI: 10.2174/011574888x311270240319084835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024]
Abstract
Exosomes secreted by mesenchymal stem/stromal cells (MSC-Exos) are advantageous candidate sources for novel acellular therapy. Despite the current standards of good manufacturing practice (GMP), the deficiency of suitable quality-control methods and the difficulties in large-scale preparation largely restrict the development of therapeutic products and their clinical applications worldwide. Herein, we mainly focus on three dominating issues commonly encountered in exosomal GMP, including issues upstream of the cell culture process, downstream of the purification process, exosomes quality control, and the drug properties of exosomes and their druggability from a corporate perspective. Collectively, in this review article, we put forward the issues of preparing clinical exosome drugs for the treatment of diverse diseases and provide new references for the clinical application of GMP-grade MSC-Exos.
Collapse
Affiliation(s)
- Fan Zhang
- Faculty of Life Sciences and Medicine, Kunming University of Science and Technology, Kunming, 650500, China
| | - Leisheng Zhang
- Science and Technology Innovation Center, The Fourth People's Hospital of Jinan (The Third Affiliated Hospital of Shandong First Medical University), Jinan, 250031, China
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hao Yu
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
26
|
St‐Denis‐Bissonnette F, Cummings SE, Qiu S, Stalker A, Muradia G, Mehic J, Mediratta K, Kaczmarek S, Burger D, Lee S, Wang L, Lavoie JR. A clinically relevant large-scale biomanufacturing workflow to produce natural killer cells and natural killer cell-derived extracellular vesicles for cancer immunotherapy. J Extracell Vesicles 2023; 12:e12387. [PMID: 38054534 PMCID: PMC10698709 DOI: 10.1002/jev2.12387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 12/07/2023] Open
Abstract
Natural killer cell-derived extracellular vesicles (NK-EVs) have shown promising potential as biotherapeutics for cancer due to their unique attributes as cytotoxic nanovesicles against cancer cells and immune-modulatory activity towards immune cells. However, a biomanufacturing workflow is needed to produce clinical-grade NK-EVs for pre-clinical and clinical applications. This study established a novel biomanufacturing workflow using a closed-loop hollow-fibre bioreactor to continuously produce NK-EVs from the clinically relevant NK92-MI cell line under serum-free, Xeno-free and feeder-free conditions following GMP-compliant conditions. The NK92 cells grown in the bioreactor for three continuous production lots resulted in large quantities of both NK cell and NK-EV biotherapeutics at the end of each production lot (over 109 viable cells and 1013 EVs), while retaining their cytotoxic payload (granzyme B and perforin), pro-inflammatory cytokine (interferon-gamma) content and cytotoxicity against the human leukemic cell line K562 with limited off-target toxicity against healthy human fibroblast cells. This scalable biomanufacturing workflow has the potential to facilitate the clinical translation of adoptive NK cell-based and NK-EV-based immunotherapies for cancer with GMP considerations.
Collapse
Affiliation(s)
- Frederic St‐Denis‐Bissonnette
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs DirectorateHealth Products and Food Branch, Health CanadaOttawaONCanada
- Department of Biochemistry, Microbiology and Immunology, Faculty of MedicineUniversity of OttawaOttawaONCanada
| | - Sarah E. Cummings
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs DirectorateHealth Products and Food Branch, Health CanadaOttawaONCanada
| | - Shirley Qiu
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs DirectorateHealth Products and Food Branch, Health CanadaOttawaONCanada
| | - Andrew Stalker
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs DirectorateHealth Products and Food Branch, Health CanadaOttawaONCanada
| | - Gauri Muradia
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs DirectorateHealth Products and Food Branch, Health CanadaOttawaONCanada
| | - Jelica Mehic
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs DirectorateHealth Products and Food Branch, Health CanadaOttawaONCanada
| | - Karan Mediratta
- Department of Biochemistry, Microbiology and Immunology, Faculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for Infection, Immunity and InflammationUniversity of OttawaOttawaONCanada
- Ottawa Institute of Systems BiologyUniversity of OttawaOttawaONCanada
| | - Shelby Kaczmarek
- Department of Biochemistry, Microbiology and Immunology, Faculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for Infection, Immunity and InflammationUniversity of OttawaOttawaONCanada
| | - Dylan Burger
- Kidney Research CentreOttawa Hospital Research InstituteOttawaONCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaONCanada
| | - Seung‐Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for Infection, Immunity and InflammationUniversity of OttawaOttawaONCanada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of MedicineUniversity of OttawaOttawaONCanada
- Centre for Infection, Immunity and InflammationUniversity of OttawaOttawaONCanada
- Ottawa Institute of Systems BiologyUniversity of OttawaOttawaONCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaONCanada
| | - Jessie R. Lavoie
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs DirectorateHealth Products and Food Branch, Health CanadaOttawaONCanada
- Department of Biochemistry, Microbiology and Immunology, Faculty of MedicineUniversity of OttawaOttawaONCanada
| |
Collapse
|
27
|
Guo J, Wang H, Li Y, Zhu S, Hu H, Gu Z. Nanotechnology in coronary heart disease. Acta Biomater 2023; 171:37-67. [PMID: 37714246 DOI: 10.1016/j.actbio.2023.09.011] [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: 05/22/2023] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Coronary heart disease (CHD) is one of the major causes of death and disability worldwide, especially in low- and middle-income countries and among older populations. Conventional diagnostic and therapeutic approaches have limitations such as low sensitivity, high cost and side effects. Nanotechnology offers promising alternative strategies for the diagnosis and treatment of CHD by exploiting the unique properties of nanomaterials. In this review, we use bibliometric analysis to identify research hotspots in the application of nanotechnology in CHD and provide a comprehensive overview of the current state of the art. Nanomaterials with enhanced imaging and biosensing capabilities can improve the early detection of CHD through advanced contrast agents and high-resolution imaging techniques. Moreover, nanomaterials can facilitate targeted drug delivery, tissue engineering and modulation of inflammation and oxidative stress, thus addressing multiple aspects of CHD pathophysiology. We discuss the application of nanotechnology in CHD diagnosis (imaging and sensors) and treatment (regulation of macrophages, cardiac repair, anti-oxidative stress), and provide insights into future research directions and clinical translation. This review serves as a valuable resource for researchers and clinicians seeking to harness the potential of nanotechnology in the management of CHD. STATEMENT OF SIGNIFICANCE: Coronary heart disease (CHD) is the one of leading cause of death and disability worldwide. Nanotechnology offers new strategies for diagnosing and treating CHD by exploiting the unique properties of nanomaterials. This review uses bibliometric analysis to uncover research trends in the use of nanotechnology for CHD. We discuss the potential of nanomaterials for early CHD detection through advanced imaging and biosensing, targeted drug delivery, tissue engineering, and modulation of inflammation and oxidative stress. We also offer insights into future research directions and potential clinical applications. This work aims to guide researchers and clinicians in leveraging nanotechnology to improve CHD patient outcomes and quality of life.
Collapse
Affiliation(s)
- Junsong Guo
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hao Wang
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Ying Li
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Houxiang Hu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| | - Zhanjun Gu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
28
|
Joo HS, Suh JH, So CM, Jeon HJ, Yoon SH, Lee JM. Emerging Roles of Using Small Extracellular Vesicles as an Anti-Cancer Drug. Int J Mol Sci 2023; 24:14063. [PMID: 37762393 PMCID: PMC10531913 DOI: 10.3390/ijms241814063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are emerging as a novel therapeutic strategy for cancer therapy. Tumor-cell-derived sEVs contain biomolecules that can be utilized for cancer diagnosis. sEVs can directly exert tumor-killing effects or modulate the tumor microenvironment, leading to anti-cancer effects. In this review, the application of sEVs as a diagnostic tool, drug delivery system, and active pharmaceutical ingredient for cancer therapy will be highlighted. The therapeutic efficacies of sEVs will be compared to conventional immune checkpoint inhibitors. Additionally, this review will provide strategies for sEV engineering to enhance the therapeutic efficacies of sEVs. As a bench-to-bedside application, we will discuss approaches to encourage good-manufacturing-practice-compliant industrial-scale manufacturing and purification of sEVs.
Collapse
Affiliation(s)
| | | | | | | | | | - Jung Min Lee
- School of Life Science, Handong Global University, 558 Handong-ro, Buk-gu, Pohang 37554, Republic of Korea
| |
Collapse
|
29
|
Costa MHG, Costa MS, Painho B, Sousa CD, Carrondo I, Oltra E, Pelacho B, Prosper F, Isidro IA, Alves P, Serra M. Enhanced bioprocess control to advance the manufacture of mesenchymal stromal cell-derived extracellular vesicles in stirred-tank bioreactors. Biotechnol Bioeng 2023; 120:2725-2741. [PMID: 36919232 DOI: 10.1002/bit.28378] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/21/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023]
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) act as signaling mediators of cellular responses. However, despite representing a promising alternative to cell-based therapies, clinical translation of EVs is currently limited by their lack of scalability and standardized bioprocessing. Herein, we integrated scalable downstream processing protocols with standardized expansion of large numbers of viable cells in stirred-tank bioreactors to improve EV production. Higher EV yields were linked to EV isolation by tangential flow filtration followed by size exclusion chromatography, rendering 5 times higher number of EVs comparatively to density gradient ultracentrifugation protocols. Additionally, when compared to static culture, EV manufacture in bioreactors resulted in 2.2 higher yields. Highlighting the role of operating under optimal cell culture conditions to maximize the number of EVs secreted per cell, MSCs cultured at lower glucose concentration favored EV secretion. While offline measurements of metabolites concentration can be performed, in this work, Raman spectroscopy was also applied to continuously track glucose levels in stirred-tank bioreactors, contributing to streamline the selection of optimal EV collection timepoints. Importantly, MSC-derived EVs retained their quality attributes and were able to stimulate angiogenesis in vitro, therefore highlighting their promising therapeutic potential.
Collapse
Affiliation(s)
- Marta H G Costa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Margarida S Costa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Beatriz Painho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Carolina D Sousa
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Inês Carrondo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Enrique Oltra
- Department of Regenerative Medicine, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Beatriz Pelacho
- Department of Regenerative Medicine, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Felipe Prosper
- Department of Regenerative Medicine, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Inês A Isidro
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Paula Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Margarida Serra
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| |
Collapse
|
30
|
Johnson J, Law SQK, Shojaee M, Hall AS, Bhuiyan S, Lim MBL, Silva A, Kong KJW, Schoppet M, Blyth C, Ranasinghe HN, Sejic N, Chuei MJ, Tatford OC, Cifuentes‐Rius A, James PF, Tester A, Dixon I, Lichtfuss G. First-in-human clinical trial of allogeneic, platelet-derived extracellular vesicles as a potential therapeutic for delayed wound healing. J Extracell Vesicles 2023; 12:e12332. [PMID: 37353884 PMCID: PMC10290200 DOI: 10.1002/jev2.12332] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/04/2023] [Accepted: 05/23/2023] [Indexed: 06/25/2023] Open
Abstract
The release of growth factors, cytokines and extracellular matrix modifiers by activated platelets is an important step in the process of healthy wound healing. Extracellular vesicles (EVs) released by activated platelets carry this bioactive cargo in an enriched form, and may therefore represent a potential therapeutic for the treatment of delayed wound healing, such as chronic wounds. While EVs show great promise in regenerative medicine, their production at clinical scale remains a critical challenge and their tolerability in humans is still to be fully established. In this work, we demonstrate that Ligand-based Exosome Affinity Purification (LEAP) chromatography can successfully isolate platelet EVs (pEVs) of clinical grade from activated platelets, which retain the regenerative properties of the parent cell. LEAP-isolated pEVs display the expected biophysical features of EV populations and transport essential proteins in wound healing processes, including insulin growth factor (IGF) and transforming growth factor beta (TGF-ß). In vitro studies show that pEVs induce proliferation and migration of dermal fibroblasts and increase dermal endothelial cells' angiogenic potential, demonstrating their wound healing potential. pEV treatment activates the ERK and Akt signalling pathways within recipient cells. In a first-in-human, double-blind, placebo-controlled, phase I clinical trial of healthy volunteer adults, designed primarily to assess safety in the context of wound healing, we demonstrate that injections of LEAP-purified pEVs in formulation buffer are safe and well tolerated (Plexoval II study, ACTRN12620000944932). As a secondary objective, biological activity in the context of wound healing rate was assessed. In this cohort of healthy participants, in which the wound bed would not be expected to be deficient in the bioactive cargo that pEVs carry, all wounds healed rapidly and completely and no difference in time to wound closure of the treated and untreated wounds was observed at the single dose tested. The outcomes of this study evidence that pEVs manufactured through the LEAP process can be injected safely in humans as a potential wound healing treatment, and warrant further study in clinical trials designed expressly to assess therapeutic efficacy in patients with delayed or disrupted wound healing.
Collapse
Affiliation(s)
- Jancy Johnson
- Exopharm LtdMelbourneVICAustralia
- Department of Biochemistry and PharmacologyUniversity of MelbourneParkvilleVICAustralia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Gregor Lichtfuss
- Exopharm LtdMelbourneVICAustralia
- Department of Biochemistry and PharmacologyUniversity of MelbourneParkvilleVICAustralia
| |
Collapse
|
31
|
Pincela Lins PM, Pirlet E, Szymonik M, Bronckaers A, Nelissen I. Manufacture of extracellular vesicles derived from mesenchymal stromal cells. Trends Biotechnol 2023; 41:965-981. [PMID: 36750391 DOI: 10.1016/j.tibtech.2023.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/09/2022] [Accepted: 01/05/2023] [Indexed: 02/08/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a promising therapy for various diseases ranging from ischemic stroke to wound healing and cancer. Their therapeutic effects are mainly mediated by secretome-derived paracrine factors, with extracellular vesicles (EVs) proven to play a key role. This has led to promising research on the potential of MSC-EVs as regenerative, off-the-shelf therapeutic agents. However, the translation of MSC-EVs into the clinic is hampered by the poor scalability of their production. Recently, new advanced methods have been developed to upscale MSC cultivation and EV production yields, ranging from new cell culture devices to priming procedures. This review gives an overview of these innovative strategies for manufacturing MSC-EVs.
Collapse
Affiliation(s)
- Paula M Pincela Lins
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium; Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium
| | - Elke Pirlet
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium
| | - Michal Szymonik
- Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium
| | - Annelies Bronckaers
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Agoralaan, 3590 Diepenbeek, Belgium.
| | - Inge Nelissen
- Flemish Institute for Technological Research (VITO), Health Department, Boeretang, 2400 Mol, Belgium.
| |
Collapse
|
32
|
Son JP, Kim EH, Shin EK, Kim DH, Sung JH, Oh MJ, Cha JM, Chopp M, Bang OY. Mesenchymal Stem Cell-Extracellular Vesicle Therapy for Stroke: Scalable Production and Imaging Biomarker Studies. Stem Cells Transl Med 2023:szad034. [PMID: 37311045 DOI: 10.1093/stcltm/szad034] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/15/2023] [Indexed: 06/15/2023] Open
Abstract
A major clinical hurdle to translate MSC-derived extracellular vesicles (EVs) is the lack of a method to scale-up the production of EVs with customized therapeutic properties. In this study, we tested whether EV production by a scalable 3D-bioprocessing method is feasible and improves neuroplasticity in animal models of stroke using MRI study. MSCs were cultured in a 3D-spheroid using a micro-patterned well. The EVs were isolated with filter and tangential flow filtration and characterized using electron microscopy, nanoparticle tracking analysis, and small RNA sequencing. Compared to conventional 2D culture, the production-reproduction of EVs (the number/size of particles and EV purity) obtained from 3D platform were more consistent among different lots from the same donor and among different donors. Several microRNAs with molecular functions associated with neurogenesis were upregulated in EVs obtained from 3D platform. EVs induced both neurogenesis and neuritogenesis via microRNAs (especially, miR-27a-3p and miR-132-3p)-mediated actions. EV therapy improved functional recovery on behavioral tests and reduced infarct volume on MRI in stroke models. The dose of MSC-EVs of 1/30 cell dose had similar therapeutic effects. In addition, the EV group had better anatomical and functional connectivity on diffusion tensor imaging and resting-state functional MRI in a mouse stroke model. This study shows that clinical-scale MSC-EV therapeutics are feasible, cost-effective, and improve functional recovery following experimental stroke, with a likely contribution from enhanced neurogenesis and neuroplasticity.
Collapse
Affiliation(s)
- Jeong Pyo Son
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup, South Korea
| | - Eun Hee Kim
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
- R&D Division, S&E bio Co., Ltd., Seoul, South Korea
| | - Eun Kyoung Shin
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
- R&D Division, S&E bio Co., Ltd., Seoul, South Korea
| | - Dong Hee Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
| | - Ji Hee Sung
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
- R&D Division, S&E bio Co., Ltd., Seoul, South Korea
| | - Mi Jeong Oh
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
| | - Jae Min Cha
- 3D Stem Cell Bioprocessing Laboratory, Department of Mechatronics, Incheon National University, Incheon, South Korea
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Oh Young Bang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
- R&D Division, S&E bio Co., Ltd., Seoul, South Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, South Korea
| |
Collapse
|
33
|
Kwak G, Gololobova O, Sharma N, Caine C, Mazur M, Mulka K, West NE, Solomon GM, Cutting GR, Witwer KW, Rowe SM, Paulaitis M, Aslanidi G, Suk JS. Extracellular vesicles enhance pulmonary transduction of stably associated adeno-associated virus following intratracheal administration. J Extracell Vesicles 2023; 12:e12324. [PMID: 37272896 PMCID: PMC10241173 DOI: 10.1002/jev2.12324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/29/2023] [Indexed: 06/06/2023] Open
Abstract
Adeno-associated virus (AAV) vector has shown multiple clinical breakthroughs, but its clinical implementation in inhaled gene therapy remains elusive due to difficulty in transducing lung airway cells. We demonstrate here AAV serotype 6 (AAV6) associated with extracellular vesicles (EVs) and secreted from vector-producing HEK-293 cells during vector preparation (EVAAV6) as a safe and highly efficacious gene delivery platform for inhaled gene therapy applications. Specifically, we discovered that EVAAV6 provided markedly enhanced reporter transgene expression in mucus-covered air-liquid interface (ALI) cultures of primary human bronchial and nasal epithelial cells as well as in mouse lung airways compared to standard preparations of AAV6 alone. Of note, AAV6 has been previously shown to outperform other clinically tested AAV serotypes, including those approved by the FDA for treating non-lung diseases, in transducing ALI cultures of primary human airway cells. We provide compelling experimental evidence that the superior performance of EVAAV6 is attributed to the ability of EV to facilitate mucus penetration and cellular entry/transduction of AAV6. The tight and stable linkage between AAV6 and EVs appears essential to exploit the benefits of EVs given that a physical mixture of individually prepared EVs and AAV6 failed to mediate EV-AAV6 interactions or to enhance gene transfer efficacy.
Collapse
Affiliation(s)
- Gijung Kwak
- Center for Nanomedicine at Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of OphthalmologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Neeraj Sharma
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Colin Caine
- Hormel InstituteUniversity of MinnesotaAustinMinnesotaUSA
| | - Marina Mazur
- Gregory Fleming James Cystic Fibrosis Research CenterHeersink School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Kathleen Mulka
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Natalie E. West
- Department of Pulmonary and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - George M. Solomon
- Gregory Fleming James Cystic Fibrosis Research CenterHeersink School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Garry R. Cutting
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Steven M. Rowe
- Gregory Fleming James Cystic Fibrosis Research CenterHeersink School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Michael Paulaitis
- Center for Nanomedicine at Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - George Aslanidi
- Hormel InstituteUniversity of MinnesotaAustinMinnesotaUSA
- Masonic Cancer CenterUniversity of MinnesotaMinneapolisMinnesotaUSA
- Institute for Molecular VirologyUniversity of MinnesotaMinneapolisUSAMinnesota
| | - Jung Soo Suk
- Center for Nanomedicine at Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of OphthalmologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
34
|
O’Brien TJ, Hollinshead F, Goodrich LR. Extracellular vesicles in the treatment and prevention of osteoarthritis: can horses help us translate this therapy to humans? EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:151-169. [PMID: 37829144 PMCID: PMC10568983 DOI: 10.20517/evcna.2023.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Osteoarthritis (OA) is a common joint disease affecting humans and horses, resulting in significant morbidity, financial expense, and loss of athletic use. While the pathogenesis is incompletely understood, inflammation is considered crucial in the development and progression of the disease. Mesenchymal stromal cells (MSCs) have received increasing scientific attention for their anti-inflammatory, immunomodulatory, and pro-regenerative effects. However, there are concerns about their ability to become a commercially available therapeutic. Extracellular vesicles (EVs) are now recognized to play a crucial role in the therapeutic efficacy observed with MSCs and offer a potentially novel cell-free therapeutic that may negate many of the concerns with MSCs. There is evidence that EVs have profound anti-inflammatory, immunomodulatory, and pro-regenerative effects equal to or greater than the MSCs they are derived from in the treatment of OA. Most of these studies are in small animal models, limiting the translation of these results to humans. However, highly translational animal models are crucial for further understanding the efficacy of potential therapeutics and for close comparisons with humans. For this reason, the horse, which experiences the same gravitational impacts on joints similar to people, is a highly relevant large animal species for testing. The equine species has well-designed and validated OA models, and additionally, therapies can be further tested in naturally occurring OA to validate preclinical model testing. Therefore, the horse is a highly suitable model to increase our knowledge of the therapeutic potential of EVs.
Collapse
Affiliation(s)
- Thomas J O’Brien
- Department of Clinical Sciences, Veterinary Teaching Hospital, Colorado State University, Fort Collins, CO 80523, USA
| | - Fiona Hollinshead
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Laurie R Goodrich
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
35
|
Wang L, Wu Y, Yao R, Li Y, Wei Y, Cao Y, Zhang Z, Wu M, Zhu H, Yao Y, Kang H. The role of mesenchymal stem cell-derived extracellular vesicles in inflammation-associated programmed cell death. NANO TODAY 2023; 50:101865. [DOI: 10.1016/j.nantod.2023.101865] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
36
|
Debnath K, Heras KL, Rivera A, Lenzini S, Shin JW. Extracellular vesicle-matrix interactions. NATURE REVIEWS. MATERIALS 2023; 8:390-402. [PMID: 38463907 PMCID: PMC10919209 DOI: 10.1038/s41578-023-00551-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 03/12/2024]
Abstract
The extracellular matrix in microenvironments harbors a variety of signals to control cellular functions and the materiality of tissues. Most efforts to synthetically reconstitute the matrix by biomaterial design have focused on decoupling cell-secreted and polymer-based cues. Cells package molecules into nanoscale lipid membrane-bound extracellular vesicles and secrete them. Thus, extracellular vesicles inherently interact with the meshwork of the extracellular matrix. In this Review, we discuss various aspects of extracellular vesicle-matrix interactions. Cells receive feedback from the extracellular matrix and leverage intracellular processes to control the biogenesis of extracellular vesicles. Once secreted, various biomolecular and biophysical factors determine whether extracellular vesicles are locally incorporated into the matrix or transported out of the matrix to be taken up by other cells or deposited into tissues at a distal location. These insights can be utilized to develop engineered biomaterials where EV release and retention can be precisely controlled in host tissue to elicit various biological and therapeutic outcomes.
Collapse
Affiliation(s)
- Koushik Debnath
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kevin Las Heras
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU)
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Ambar Rivera
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Stephen Lenzini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
37
|
Steć A, Chodkowska M, Kasprzyk-Pochopień J, Mielczarek P, Piekoszewski W, Lewczuk B, Płoska A, Kalinowski L, Wielgomas B, Dziomba S. Isolation of Citrus lemon extracellular vesicles: Development and process control using capillary electrophoresis. Food Chem 2023; 424:136333. [PMID: 37201469 DOI: 10.1016/j.foodchem.2023.136333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023]
Abstract
A new and scalable method for the isolation of extracellular vesicles (EV) from Citrus lemon juice samples was developed. The methodology included preliminary preconcentration of the sample using ultrafiltration (UF) followed by size-exclusion chromatography (SEC) purification and final preconcentration of the eluates. Transmission electron microscopy and proteomic analysis showed that isolates contained exosome-like vesicles, exocyst-positive organelle (EXPO), and microvesicles. The efficiency of certain isolation steps was evaluated with total protein content assay (bicinchoninic acid assay, BCA), nanoparticles tracking analysis (NTA), and capillary electrophoresis (CE). A good correlation between CE, BCA, and NTA results was shown. The application of CE enabled the detection of soluble contaminants, macromolecular aggregates, and vesicles' heterogeneity. The fluorescent staining of encapsulated nucleic acids was proposed for the identity confirmation of EV detected in CE. The study demonstrates the CE as a comprehensive tool for monitoring of the EV isolation process.
Collapse
Affiliation(s)
- Aleksandra Steć
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland
| | - Martyna Chodkowska
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland
| | - Joanna Kasprzyk-Pochopień
- Laboratory of High-Resolution Mass Spectrometry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland
| | - Przemyslaw Mielczarek
- AGH University of Science and Technology, Faculty of Materials Science and Ceramics, Department of Analytical Chemistry and Biochemistry, 30 Mickiewicza Avenue, 30-059 Krakow, Poland
| | - Wojciech Piekoszewski
- Laboratory of High-Resolution Mass Spectrometry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland; Department of Analytical Chemistry, Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Street, 30-387 Krakow, Poland
| | - Bogdan Lewczuk
- University of Warmia and Mazury in Olsztyn, Department of Histology and Embryology, 13 Oczapowskiego Street, 10-713 Olsztyn, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland; BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland
| | - Bartosz Wielgomas
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland
| | - Szymon Dziomba
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, 107 Hallera Street, 80-416 Gdansk, Poland.
| |
Collapse
|
38
|
Nicodemou A, Bernátová S, Čeháková M, Danišovič Ľ. Emerging Roles of Mesenchymal Stem/Stromal-Cell-Derived Extracellular Vesicles in Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15051453. [PMID: 37242693 DOI: 10.3390/pharmaceutics15051453] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Despite the tremendous efforts of many researchers and clinicians, cancer remains the second leading cause of mortality worldwide. Mesenchymal stem/stromal cells (MSCs) are multipotent cells residing in numerous human tissues and presenting unique biological properties, such as low immunogenicity, powerful immunomodulatory and immunosuppressive capabilities, and, in particular, homing abilities. Therapeutic functions of MSCs are mediated mostly by the paracrine effect of released functional molecules and other variable components, and among them the MSC-derived extracellular vesicles (MSC-EVs) seem to be one of the central mediators of the therapeutic functions of MSCs. MSC-EVs are membrane structures secreted by the MSCs, rich in specific proteins, lipids, and nucleic acids. Amongst these, microRNAs have achieved the most attention currently. Unmodified MSC-EVs can promote or inhibit tumor growth, while modified MSC-EVs are involved in the suppression of cancer progression via the delivery of therapeutic molecules, including miRNAs, specific siRNAs, or suicide RNAs, as well as chemotherapeutic drugs. Here, we present an overview of the characteristics of the MSCs-EVs and describe the current methods for their isolation and analysis, the content of their cargo, and modalities for the modification of MSC-EVs in order for them to be used as drug delivery vehicles. Finally, we describe different roles of MSC-EVs in the tumor microenvironment and summarize current advances of MCS-EVs in cancer research and therapy. MSC-EVs are expected to be a novel and promising cell-free therapeutic drug delivery vehicle for the treatment of cancer.
Collapse
Affiliation(s)
- Andreas Nicodemou
- Lambda Life a. s., Levocska 3617/3, 851 01 Bratislava, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Soňa Bernátová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Michaela Čeháková
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľuboš Danišovič
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Centre for Tissue Engineering and Regenerative Medicine-Translational Research Unit in the Branch of Regenerative Medicine, Faculty of Medicine, Comenius University, Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
39
|
Zhong Y, Wang X, Zhao X, Shen J, Wu X, Gao P, Yang P, Chen J, An W. Multifunctional Milk-Derived Small Extracellular Vesicles and Their Biomedical Applications. Pharmaceutics 2023; 15:1418. [PMID: 37242660 PMCID: PMC10223436 DOI: 10.3390/pharmaceutics15051418] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
In recent years, small extracellular vesicles (sEVs) have been regarded as the next generation of novel delivery systems after lipid nanoparticles because of their advantages and huge prospects in drug delivery. Studies have shown that sEVs are abundant in milk and therefore can be a large and economical source of sEVs. Natural milk-derived small extracellular vesicles (msEVs) have important functions such as immune regulation, anti-bacterial infection, anti-oxidative, etc., and play a beneficial role in human health at multiple levels, including intestinal health, bone/muscle metabolism, and microbiota regulation. In addition, because they can pass the gastrointestinal barrier and have low immunogenicity, good biocompatibility, and stability, msEVs are considered a crucial oral drug delivery vehicle. Moreover, msEVs can be further engineered for targeted delivery to prolong the circulation time or enhance local drug concentrations. However, msEVs separation and purification, complex contents, and quality control hinder their application in drug delivery. This paper provides a comprehensive review of the biogenesis and characteristics, isolation and purification, composition, loading methods, and function of msEVs, based on which their applications in biomedical fields are further explored.
Collapse
Affiliation(s)
- Youxiu Zhong
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xudong Wang
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xian Zhao
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Jiuheng Shen
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xue Wu
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Peifen Gao
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Peng Yang
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 100083, China
| | - Wenlin An
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| |
Collapse
|
40
|
Brezgin S, Parodi A, Kostyusheva A, Ponomareva N, Lukashev A, Sokolova D, Pokrovsky VS, Slatinskaya O, Maksimov G, Zamyatnin AA, Chulanov V, Kostyushev D. Technological aspects of manufacturing and analytical control of biological nanoparticles. Biotechnol Adv 2023; 64:108122. [PMID: 36813011 DOI: 10.1016/j.biotechadv.2023.108122] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/19/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived biological nanoparticles that gained great interest for drug delivery. EVs have numerous advantages compared to synthetic nanoparticles, such as ideal biocompatibility, safety, ability to cross biological barriers and surface modification via genetic or chemical methods. On the other hand, the translation and the study of these carriers resulted difficult, mostly because of significant issues in up-scaling, synthesis and impractical methods of quality control. However, current manufacturing advances enable EV packaging with any therapeutic cargo, including DNA, RNA (for RNA vaccines and RNA therapeutics), proteins, peptides, RNA-protein complexes (including gene-editing complexes) and small molecules drugs. To date, an array of new and upgraded technologies have been introduced, substantially improving EV production, isolation, characterization and standardization. The used-to-be "gold standards" of EV manufacturing are now outdated, and the state-of-art requires extensive revision. This review re-evaluates the pipeline for EV industrial production and provides a critical overview of the modern technologies required for their synthesis and characterization.
Collapse
Affiliation(s)
- Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia
| | | | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia
| | - Darina Sokolova
- Sirius University of Science and Technology, Sochi 354340, Russia; Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; People's Friendship University, Moscow 117198, Russia
| | - Vadim S Pokrovsky
- Sirius University of Science and Technology, Sochi 354340, Russia; Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; People's Friendship University, Moscow 117198, Russia
| | - Olga Slatinskaya
- Lomonosov Moscow State University, Faculty of Biology, Moscow 119991, Russia
| | - Georgy Maksimov
- Lomonosov Moscow State University, Faculty of Biology, Moscow 119991, Russia
| | - Andrey A Zamyatnin
- Sirius University of Science and Technology, Sochi 354340, Russia; Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7X, UK
| | - Vladimir Chulanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia; Department of Infectious Diseases, Sechenov University, Moscow 119048, Russia; National Medical Research Center for Tuberculosis and Infectious Diseases, Moscow 127994, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow 119048, Russia; Sirius University of Science and Technology, Sochi 354340, Russia.
| |
Collapse
|
41
|
Thompson W, Papoutsakis ET. The role of biomechanical stress in extracellular vesicle formation, composition and activity. Biotechnol Adv 2023; 66:108158. [PMID: 37105240 DOI: 10.1016/j.biotechadv.2023.108158] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Extracellular vesicles (EVs) are cornerstones of intercellular communication with exciting fundamental, clinical, and more broadly biotechnological applications. However, variability in EV composition, which results from the culture conditions used to generate the EVs, poses significant fundamental and applied challenges and a hurdle for scalable bioprocessing. Thus, an understanding of the relationship between EV production (and for clinical applications, manufacturing) and EV composition is increasingly recognized as important and necessary. While chemical stimulation and culture conditions such as cell density are known to influence EV biology, the impact of biomechanical forces on the generation, properties, and biological activity of EVs remains poorly understood. Given the omnipresence of these forces in EV preparation and in biomanufacturing, expanding the understanding of their impact on EV composition-and thus, activity-is vital. Although several publications have examined EV preparation and bioprocessing and briefly discussed biomechanical stresses as variables of interest, this review represents the first comprehensive evaluation of the impact of such stresses on EV production, composition and biological activity. We review how EV biogenesis, cargo, efficacy, and uptake are uniquely affected by various types, magnitudes, and durations of biomechanical forces, identifying trends that emerge both generically and for individual cell types. We also describe implications for scalable bioprocessing, evaluating processes inherent in common EV production and isolation methods, and propose a path forward for rigorous EV quality control.
Collapse
Affiliation(s)
- Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA.
| |
Collapse
|
42
|
Pan W, Chen H, Wang A, Wang F, Zhang X. Challenges and strategies: Scalable and efficient production of mesenchymal stem cells-derived exosomes for cell-free therapy. Life Sci 2023; 319:121524. [PMID: 36828131 DOI: 10.1016/j.lfs.2023.121524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/10/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023]
Abstract
Exosomes are small membrane vesicles secreted by most cell types, and widely exist in cell supernatants and various body fluids. They can transmit numerous bioactive elements, such as proteins, nucleic acids, and lipids, to affect the gene expression and function of recipient cells. Mesenchymal stem cells (MSCs) have been confirmed to be a potentially promising therapy for tissue repair and regeneration. Accumulating studies demonstrated that the predominant regenerative paradigm of MSCs transplantation was the paracrine effect but not the differentiation effect. Exosomes secreted by MSCs also showed similar therapeutic effects as their parent cells and were considered to be used for cell-free regenerative medicine. However, the inefficient and limited production has hampered their development for clinical translation. In this review, we summarize potential methods to efficiently promote the yield of exosomes. We mainly focus on engineering the process of exosome biogenesis and secretion, altering the cell culture conditions, cell expansion through 3D dynamic culture and the isolation of exosomes. In addition, we also discuss the application of MSCs-derived exosomes as therapeutics in disease treatment.
Collapse
Affiliation(s)
- Wei Pan
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongyuan Chen
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 Jingwuweiqi Road 324, Jinan 250021, China
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, UC Davis Health Medical Center, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Fengshan Wang
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; National Glycoengineering Research Center, Shandong University, Jinan, Shandong 250012, China.
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
43
|
Kalluri R, McAndrews KM. The role of extracellular vesicles in cancer. Cell 2023; 186:1610-1626. [PMID: 37059067 PMCID: PMC10484374 DOI: 10.1016/j.cell.2023.03.010] [Citation(s) in RCA: 201] [Impact Index Per Article: 100.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 04/16/2023]
Abstract
Intercellular communication is a key feature of cancer progression and metastasis. Extracellular vesicles (EVs) are generated by all cells, including cancer cells, and recent studies have identified EVs as key mediators of cell-cell communication via packaging and transfer of bioactive constituents to impact the biology and function of cancer cells and cells of the tumor microenvironment. Here, we review recent advances in understanding the functional contribution of EVs to cancer progression and metastasis, as cancer biomarkers, and the development of cancer therapeutics.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Kathleen M McAndrews
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
44
|
Chan AML, Cheah JM, Lokanathan Y, Ng MH, Law JX. Natural Killer Cell-Derived Extracellular Vesicles as a Promising Immunotherapeutic Strategy for Cancer: A Systematic Review. Int J Mol Sci 2023; 24:ijms24044026. [PMID: 36835438 PMCID: PMC9964266 DOI: 10.3390/ijms24044026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
Cancer is the second leading contributor to global deaths caused by non-communicable diseases. The cancer cells are known to interact with the surrounding non-cancerous cells, including the immune cells and stromal cells, within the tumor microenvironment (TME) to modulate the tumor progression, metastasis and resistance. Currently, chemotherapy and radiotherapy are the standard treatments for cancers. However, these treatments cause a significant number of side effects, as they damage both the cancer cells and the actively dividing normal cells indiscriminately. Hence, a new generation of immunotherapy using natural killer (NK) cells, cytotoxic CD8+ T-lymphocytes or macrophages was developed to achieve tumor-specific targeting and circumvent the adverse effects. However, the progression of cell-based immunotherapy is hindered by the combined action of TME and TD-EVs, which render the cancer cells less immunogenic. Recently, there has been an increase in interest in using immune cell derivatives to treat cancers. One of the highly potential immune cell derivatives is the NK cell-derived EVs (NK-EVs). As an acellular product, NK-EVs are resistant to the influence of TME and TD-EVs, and can be designed for "off-the-shelf" use. In this systematic review, we examine the safety and efficacy of NK-EVs to treat various cancers in vitro and in vivo.
Collapse
Affiliation(s)
- Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/22, Petaling Jaya 47101, Malaysia
| | - Jin Min Cheah
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/22, Petaling Jaya 47101, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Correspondence: ; Tel.: +60-391-457677
| |
Collapse
|
45
|
Tian MY, Hao DX, Liu Y, He J, Zhao ZH, Guo TY, Li X, Zhang Y. Milk exosomes: an oral drug delivery system with great application potential. Food Funct 2023; 14:1320-1337. [PMID: 36722924 DOI: 10.1039/d2fo02013k] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Exosomes are extracellular vesicles with the smallest diameter, usually divided into cellular sources and body fluid sources. Due to their special properties different from cell-derived exosomes, the application of milk exosomes as an oral drug delivery system has increased greatly. This article introduces the physical and chemical properties of exosomes, separation technology, dyeing and labeling technology, targeted modification technology, and the application of milk exosomes in drug loading and disease therapies.
Collapse
Affiliation(s)
- Meng-Yuan Tian
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Dong-Xia Hao
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Yang Liu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Jin He
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Zhuo-Hua Zhao
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Ting-Yu Guo
- The International Department of the High School Affiliated to Shaanxi Normal University, Xi'an, China
| | - Xing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Yuan Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
46
|
Williams S, Jalal AR, Lewis MP, Davies OG. A survey to evaluate parameters governing the selection and application of extracellular vesicle isolation methods. J Tissue Eng 2023; 14:20417314231155114. [PMID: 36911574 PMCID: PMC9996742 DOI: 10.1177/20417314231155114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/19/2023] [Indexed: 03/11/2023] Open
Abstract
Extracellular vesicles (EVs) continue to gain interest across the scientific community for diagnostic and therapeutic applications. As EV applications diversify, it is essential that researchers are aware of challenges, in particular the compatibility of EV isolation methods with downstream applications and their clinical translation. We report outcomes of the first cross-comparison study looking to determine parameters (EV source, starting volume, operator experience, application and implementation parameters such as cost and scalability) governing the selection of popular EV isolation methods across disciplines. Our findings highlighted an increased clinical focus, with 36% of respondents applying EVs in therapeutics and diagnostics. Data indicated preferential selection of ultracentrifugation for therapeutic applications, precipitation reagents in clinical settings and size exclusion chromatography for diagnostic applications utilising biofluids. Method selection was influenced by operator experience, with increased method diversity when EV research was not the respondents primary focus. Application and implementation criteria were indicated to be major influencers in method selection, with UC and SEC chosen for their abilities to process large and small volumes, respectively. Overall, we identified parameters influencing method selection across the breadth of EV science, providing a valuable overview of practical considerations for the effective translation of research outcomes.
Collapse
Affiliation(s)
- Soraya Williams
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Aveen R Jalal
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Owen G Davies
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
47
|
Cai R, Wang L, Zhang W, Liu B, Wu Y, Pang J, Ma C. The role of extracellular vesicles in periodontitis: pathogenesis, diagnosis, and therapy. Front Immunol 2023; 14:1151322. [PMID: 37114060 PMCID: PMC10126335 DOI: 10.3389/fimmu.2023.1151322] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Periodontitis is a prevalent disease and one of the leading causes of tooth loss. Biofilms are initiating factor of periodontitis, which can destroy periodontal tissue by producing virulence factors. The overactivated host immune response is the primary cause of periodontitis. The clinical examination of periodontal tissues and the patient's medical history are the mainstays of periodontitis diagnosis. However, there is a lack of molecular biomarkers that can be used to identify and predict periodontitis activity precisely. Non-surgical and surgical treatments are currently available for periodontitis, although both have drawbacks. In clinical practice, achieving the ideal therapeutic effect remains a challenge. Studies have revealed that bacteria produce extracellular vesicles (EVs) to export virulence proteins to host cells. Meanwhile, periodontal tissue cells and immune cells produce EVs that have pro- or anti-inflammatory effects. Accordingly, EVs play a critical role in the pathogenesis of periodontitis. Recent studies have also presented that the content and composition of EVs in saliva and gingival crevicular fluid (GCF) can serve as possible periodontitis diagnostic indicators. In addition, studies have indicated that stem cell EVs may encourage periodontal regeneration. In this article, we mainly review the role of EVs in the pathogenesis of periodontitis and discuss their diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Rong Cai
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wei Zhang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Bing Liu
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianliang Pang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| | - Chufan Ma
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| |
Collapse
|
48
|
Castillo-Romero KF, Santacruz A, González-Valdez J. Production and purification of bacterial membrane vesicles for biotechnology applications: Challenges and opportunities. Electrophoresis 2023; 44:107-124. [PMID: 36398478 DOI: 10.1002/elps.202200133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/17/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
Bacterial membrane vesicles (BMVs) are bi-layered nanostructures derived from Gram-negative and Gram-positive bacteria. Among other pathophysiological roles, BMVs are critical messengers in intercellular communication. As a result, BMVs are emerging as a promising technology for the development of numerous therapeutic applications. Despite the remarkable progress in unveiling BMV biology and functions in recent years, their successful isolation and purification have been limited. Several challenges related to vesicle purity, yield, and scalability severely hamper the further development of BMVs for biotechnology and clinical applications. This review focuses on the current technologies and methodologies used in BMV production and purification, such as ultracentrifugation, density-gradient centrifugation, size-exclusion chromatography, ultrafiltration, and precipitation. We also discuss the current challenges related to BMV isolation, large-scale production, storage, and stability that limit their application. More importantly, the present work explains the most recent strategies proposed for overcoming those challenges. Finally, we summarize the ongoing applications of BMVs in the biotechnological field.
Collapse
Affiliation(s)
- Keshia F Castillo-Romero
- School of Engineering and Science, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo León, Mexico
| | - Arlette Santacruz
- School of Engineering and Science, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo León, Mexico
| | - José González-Valdez
- School of Engineering and Science, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo León, Mexico
| |
Collapse
|
49
|
Hu P, Ying J, Wang Y, Jiang T, Pan Z, Zhao C, Li J, Li C. Extracellular Vesicles Derived From 3D Cultured Antler Stem Cells Serve as a New Drug Vehicle in Osteosarcoma Treatment. Cell Transplant 2023; 32:9636897231219830. [PMID: 38102784 PMCID: PMC10725652 DOI: 10.1177/09636897231219830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/23/2023] [Accepted: 11/26/2023] [Indexed: 12/17/2023] Open
Abstract
Extracellular vesicles (EVs) from antler reserve mesenchymal (RM) cells play an important role in the paracrine regulation during rapid growth of antler without forming a tumor; therefore, RM-EVs become novel materials for anti-tumor studies, such as osteosarcoma treatment. However, the problem of low production of RM-EVs in traditional 2D culture limits its mechanism research and application. In this study, we established an optimal 3D culture system for antler RM cells to produce EVs (3D-RM-EVs). Morphology and property of harvested 3D-RM-EVs were normal compared with EVs from conventional 2D culture, and the miRNA profile in them was basically the same through transcriptome sequencing analysis. Based on the same number of RM cells, the volume of the culture medium collected by 3D cultural system concentrated nearly 30 times, making it more convenient for subsequent purification. In addition, EVs were harvested 30 times in 3D cultural system, greatly increasing the total amount of EVs (harvested a total of 2-3 times in 2D culture). Although 3D-RM-EVs had a limited inhibitory effect on the proliferation of K7M2 cells, the inhibition effect of 3D-RM-EVs loaded drugs (Ifosfamide + Etoposide) were more significant than that of positive drug group alone (P < 0.05). Furthermore, in vivo studies showed that 3D-RM-EVs loaded drugs (Ifosfamide + Etoposide) had the most significant tumor inhibition effect, with decreased tumor size, and could slow down body weight loss compared with Ifosfamide + Etoposide (IFO + ET) group. These results demonstrated that 3D-RM-EVs were efficiently prepared from antler RM cells and were effective as drug vehicles for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Pengfei Hu
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Jinchi Ying
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Yusu Wang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Tiantian Jiang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Zheng Pan
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Chen Zhao
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Jiping Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| |
Collapse
|
50
|
Qu S, Han Y, Liu Y, Zhu J, Acaroz U, Shen J, Zhu K. Milk Exosomes Facilitate Oral Delivery of Drugs against Intestinal Bacterial Infections. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:16069-16079. [PMID: 36515136 DOI: 10.1021/acs.jafc.2c04971] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Biopharmaceutics Classification System (BCS) class II and IV drugs exhibit low solubility and suffer a limitation in oral administration. Exosomes have attracted intensive attention in the efficient delivery of such compounds. However, low gastrointestinal stability and high production cost of exosomes hinder their development as drug carriers. Here, milk exosomes are functionalized with phosphatidylserine and are capable of improving the solubility of BCS class II and IV drugs, resulting in facilitating the oral delivery of the drugs. A natural flavonoid, α-mangostin, is loaded into exosomes (AExo) to enhance the antibacterial efficiency, demonstrated by clearing 99% of bacteria in macrophages. Furthermore, AExo exhibits high mucus penetrability and shows a significant therapeutic efficacy in two animal infection models. Collectively, this work expands the application of exosomes from bovine milk with simple operation and low cost, shedding light on the potential of milk exosomes in improving the solubility of drugs to enhance the efficacy of oral administration.
Collapse
Affiliation(s)
- Shaoqi Qu
- Key Laboratory of Traditional Chinese Veterinary Medicine Biology, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Yiming Han
- College of Engineering, Peking University, Beijing 100871, China
| | - Ying Liu
- Key Laboratory of Traditional Chinese Veterinary Medicine Biology, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiajia Zhu
- Key Laboratory of Traditional Chinese Veterinary Medicine Biology, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Ulas Acaroz
- Department of Food Hygiene and Technology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar 03200, Turkey
| | - Jianzhong Shen
- Key Laboratory of Traditional Chinese Veterinary Medicine Biology, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Kui Zhu
- Key Laboratory of Traditional Chinese Veterinary Medicine Biology, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| |
Collapse
|