1
|
Yang H, Yang H, Wang Q, Ji H, Qian T, Qiao Y, Shi J, Cong M. Mesenchymal stem cells and their extracellular vesicles: new therapies for cartilage repair. Front Bioeng Biotechnol 2025; 13:1591400. [PMID: 40343207 PMCID: PMC12058886 DOI: 10.3389/fbioe.2025.1591400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
Cartilage is crucial for joints, and its damage can lead to pain and functional impairment, causing financial burden to patients. Due to its weak self-repair, cartilage injury control is a research focus. Cartilage injury naturally with age, but mechanical trauma, lifestyle factors and certain genetic abnormalities can increase the likelihood of symptomatic disease progression. Current treatments for cartilage injury include pharmacological and surgical interventions, but these lack the ability to stop the progression of disease and restore the regeneration of the cartilage. Biological therapies have been evaluated but show varying degrees of efficacy in cartilage regeneration long-term. The mesenchymal stem cell (MSC) therapy attracts attention as it is easily harvested and expanded. Once thought to repair via differentiation, MSCs are now known to secrete extracellular vesicles (EVs) paracrinely. These EVs, rich in bioactive molecules, enable cell communication, boost growth factor secretion, regulate the synthesis and degradation of extracellular matrix (ECM), and modulate inflammation, vital for cartilage repair. However, further research and clinical validation are still required for the application of MSC and MSC-EVs. This review highlights the current state of research on the use of MSC and MSC-EVs in the treatment of cartilage injury. It is hoped that the review in this paper will provide valuable references and inspiration for future researchers in therapeutic studies of cartilage repair.
Collapse
Affiliation(s)
- Hongwei Yang
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Haochen Yang
- School of Medicine, Nantong University, Nantong, China
| | - Qin Wang
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Hanzhen Ji
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Tianmei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Engineering Research Center of Integration and Application of Digital Learning Technology, Ministry of Education, Beijing, China
| | - Yusen Qiao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfeng Shi
- Department of Orthopedics, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Meng Cong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
2
|
Yoo D, Jung SY, Go D, Park JY, You DG, Jung WK, Li Y, Ding J, Park JH, Um W. Functionalized extracellular vesicles of mesenchymal stem cells for regenerative medicine. J Nanobiotechnology 2025; 23:219. [PMID: 40102934 PMCID: PMC11921732 DOI: 10.1186/s12951-025-03300-6] [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: 06/09/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Stem cell-derived extracellular vesicles (EVs) have emerged as a safe and potent alternative to regenerative medicine in recent decades. Furthermore, the adjustment of EV functions has been recently enabled by certain stem cell preconditioning methods, providing an exceptional opportunity to enhance the therapeutic potential or confer additional functions of stem cell-derived EVs. In this review, we discuss the recent progress of functionalized EVs, based on stem cell preconditioning, for treating various organ systems, such as the musculoskeletal system, nervous system, integumentary system, cardiovascular system, renal system, and respiratory system. Additionally, we summarize the expected outcomes of preconditioning methods for stem cells and their EVs. With recent progress, we suggest considerations and future directions for developing personalized medicine based on preconditioned stem cell-derived EVs.
Collapse
Affiliation(s)
- Donghyeon Yoo
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Se Young Jung
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dabin Go
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Ji Yeong Park
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dong Gil You
- Department of Chemical Engineering & Biotechnology, Tech University of Korea, Siheung, 15073, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Republic of Korea
| | - Yuce Li
- College of Life Science and Health, Wuhan University of Science and Technology (WUST), Wuhan, 430065, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
3
|
Qiu D, Yan B, Xue H, Xu Z, Tan G, Liu Y. Perspectives of exosomal ncRNAs in the treatment of bone metabolic diseases: Focusing on osteoporosis, osteoarthritis, and rheumatoid arthritis. Exp Cell Res 2025; 446:114457. [PMID: 39986599 DOI: 10.1016/j.yexcr.2025.114457] [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: 10/23/2024] [Revised: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Bone metabolic disorders, constituting a group of prevalent and grave conditions, currently have a scarcity of therapeutic alternatives. Over the recent past, exosomes have been at the forefront of research interest, owing to their nanoparticulate nature and potential for therapeutic intervention. ncRNAs are a class of heterogeneous transcripts that they lack protein-encoding capacity, yet they can modulate the expression of other genes through multiple mechanisms. Mounting evidence underscores the intricate role of exosomes as ncRNAs couriers implicated in the pathogenesis of bone metabolic disorders. In this review, we endeavor to elucidate recent insights into the roles of three ncRNAs - miRNAs, lncRNAs, and circRNAs - in bone metabolic ailments such as osteoporosis, osteoarthritis, and rheumatoid arthritis. Additionally, we examine the viability of exosomal ncRNAs as innovative, cell-free modalities in the diagnosis and therapeutic management of bone metabolic disorders. We aim to uncover the critical function of exosomal ncRNAs within the context of bone metabolic diseases.
Collapse
Affiliation(s)
- Daodi Qiu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Binghan Yan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Haipeng Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhanwang Xu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Guoqing Tan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yajuan Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250300, China.
| |
Collapse
|
4
|
Longfei H, Wenyuan H, Weihua F, Peng P, Sun L, Kun L, Mincong H, Fan Y, Wei H, Qiushi W. Exosomes in cartilage microenvironment regulation and cartilage repair. Front Cell Dev Biol 2025; 13:1460416. [PMID: 40109360 PMCID: PMC11919854 DOI: 10.3389/fcell.2025.1460416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Osteoarthritis (OA) is a debilitating disease that predominantly impacts the hip, hand, and knee joints. Its pathology is defined by the progressive degradation of articular cartilage, formation of bone spurs, and synovial inflammation, resulting in pain, joint function limitations, and substantial societal and familial burdens. Current treatment strategies primarily target pain alleviation, yet improved interventions addressing the underlying disease pathology are scarce. Recently, exosomes have emerged as a subject of growing interest in OA therapy. Numerous studies have investigated exosomes to offer promising therapeutic approaches for OA through diverse in vivo and in vitro models, elucidating the mechanisms by which exosomes from various cell sources modulate the cartilage microenvironment and promote cartilage repair. Preclinical investigations have demonstrated the regulatory effects of exosomes originating from human cells, including mesenchymal stem cells (MSC), synovial fibroblasts, chondrocytes, macrophages, and exosomes derived from Chinese herbal medicines, on the modulation of the cartilage microenvironment and cartilage repair through diverse signaling pathways. Additionally, therapeutic mechanisms encompass cartilage inflammation, degradation of the cartilage matrix, proliferation and migration of chondrocytes, autophagy, apoptosis, and mitigation of oxidative stress. An increasing number of exosome carrier scaffolds are under development. Our review adopts a multidimensional approach to enhance comprehension of the pivotal therapeutic functions exerted by exosomes sourced from diverse cell types in OA. Ultimately, our aim is to pinpoint therapeutic targets capable of regulating the cartilage microenvironment and facilitating cartilage repair in OA.
Collapse
Affiliation(s)
- Han Longfei
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hou Wenyuan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fang Weihua
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Peng Peng
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lu Sun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Kun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Mincong
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yang Fan
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Wei
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Qiushi
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Orthopaedic, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Xu H, Wang Z, Wang Z, Chen J, Zhao C, Kang B, Xu X, Shen J, Li M, Diao J, Xie J, Xiao L. Mapping the knowledge landscape: A bibliometric analysis of exosome research in osteoarthritis (2004-2023). Heliyon 2024; 10:e40079. [PMID: 39717577 PMCID: PMC11665376 DOI: 10.1016/j.heliyon.2024.e40079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/17/2024] [Accepted: 10/31/2024] [Indexed: 12/25/2024] Open
Abstract
Exosomes have emerged as a crucial focus in advancing the diagnosis and treatment of osteoarthritis (OA). However, there are limited bibliometric studies on this topic. This study aimed to delineate the literature landscape on exosomes in OA, identifying global research trends and key areas. We utilised the Web of Science Core Collection to retrieve articles published from 2004 to 2023. Our analysis included 456 publications across 671 institutions from 40 countries/regions. Publication volume, citations, and emerging research foci and trends were examined. Our results reveal a consistently increased interest in exosomes related to OA over the past two decades. Prominent institutions contributing to this research include Shanghai Jiao Tong University and Shenzhen University. The leading journal for these publications is the International Journal of Molecular Sciences, with Stem Cell Research & Therapy being the most frequently co-cited journal. Notable scholars in this field are Li Duan, Yujie Liang, Xiao Xu, and Wei Seong Toh, with Shipin Zhang emerging as the most co-cited author. The principal research themes were elucidating how exosomes contribute to OA pathology and developing novel therapeutic approaches. Research hotspots and new trends are linked to terms such as "cartilage," "mesenchymal stem cell," "miRNA," "treatment," and "biomarkers." This comprehensive analysis offers valuable insights into the prevailing scientific discourse, pivotal topics, and potential future directions. It could serve as a foundational reference for researchers exploring exosomes and their utility in OA diagnostics and therapeutics.
Collapse
Affiliation(s)
- Hui Xu
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhen Wang
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zheng Wang
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Juntao Chen
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
| | - Chi Zhao
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
| | - Bingxin Kang
- Rehabilitation Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xirui Xu
- Guanghua School of Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Joint Surgery Department, Shanghai Guanghua Integrated Chinese and Western Medicine Hospital, Shanghai, China
| | - Jun Shen
- Guanghua School of Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Joint Surgery Department, Shanghai Guanghua Integrated Chinese and Western Medicine Hospital, Shanghai, China
| | - Mengmeng Li
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jieyao Diao
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jun Xie
- Guanghua School of Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Joint Surgery Department, Shanghai Guanghua Integrated Chinese and Western Medicine Hospital, Shanghai, China
| | - Lianbo Xiao
- Guanghua School of Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Joint Surgery Department, Shanghai Guanghua Integrated Chinese and Western Medicine Hospital, Shanghai, China
| |
Collapse
|
6
|
Jin P, Liu H, Chen X, Liu W, Jiang T. From Bench to Bedside: The Role of Extracellular Vesicles in Cartilage Injury Treatment. Biomater Res 2024; 28:0110. [PMID: 39583872 PMCID: PMC11582190 DOI: 10.34133/bmr.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024] Open
Abstract
Cartilage repair is the key to the treatment of joint-related injury. However, because cartilage lacks vessels and nerves, its self-repair ability is extremely low. Extracellular vesicles (EVs) are bilayer nanovesicles with membranes mainly composed of ceramides, cholesterol, phosphoglycerides, and long-chain free fatty acids, containing DNA, RNA, and proteins (such as integrins and enzymes). For mediating intercellular communication and regulating mechanisms, EVs have been shown by multiple studies to be effective treatment options for cartilage repair. This review summarizes recent findings of different sources (mammals, plants, and bacteria) and uses of EVs in cartilage repair, mechanisms of EVs captured by injured chondrocytes, and quantification and storage of EVs, which may provide scientific guidance for promoting the development of EVs in the field of cartilage injury treatment.
Collapse
Affiliation(s)
- Pan Jin
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Huan Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Xichi Chen
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Wei Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Tongmeng Jiang
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University,
Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma; Hainan Provincial Stem Cell Research Institute; Hainan Academy of Medical Sciences,
Hainan Medical University, Haikou 571199, China
| |
Collapse
|
7
|
Selvadoss A, Baby HM, Zhang H, Bajpayee AG. Harnessing exosomes for advanced osteoarthritis therapy. NANOSCALE 2024; 16:19174-19191. [PMID: 39323205 PMCID: PMC11799831 DOI: 10.1039/d4nr02792b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
Exosomes are nanosized, lipid membrane vesicles secreted by cells, facilitating intercellular communication by transferring cargo from parent to recipient cells. This capability enables biological crosstalk across multiple tissues and cells. Extensive research has been conducted on their role in the pathogenesis of degenerative musculoskeletal diseases such as osteoarthritis (OA), a chronic and painful joint disease that particularly affects cartilage. Currently, no effective treatment exists for OA. Given that exosomes naturally modulate synovial joint inflammation and facilitate cartilage matrix synthesis, they are promising candidates as next generation nanocarriers for OA therapy. Recent advancements have focused on engineering exosomes through endogenous and exogenous approaches to enhance their joint retention, cartilage and chondrocyte targeting properties, and therapeutic content enrichment, further increasing their potential for OA drug delivery. Notably, charge-reversed exosomes that utilize electrostatic binding interactions with cartilage anionic aggrecan glycosaminoglycans have demonstrated the ability to penetrate the full thickness of early-stage arthritic cartilage tissue following intra-articular administration, maximizing their therapeutic potential. These exosomes offer a non-viral, naturally derived, cell-free carrier for OA drug and gene delivery applications. Efforts to standardize exosome harvest, engineering, and property characterization methods, along with scaling up production, will facilitate more efficient and rapid clinical translation. This article reviews the current state-of-the-art, explores opportunities for exosomes as OA therapeutics, and identifies potential challenges in their clinical translation.
Collapse
Affiliation(s)
- Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Ambika G Bajpayee
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
8
|
Abid AI, Conzatti G, Toti F, Anton N, Vandamme T. Mesenchymal stem cell-derived exosomes as cell free nanotherapeutics and nanocarriers. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 61:102769. [PMID: 38914247 DOI: 10.1016/j.nano.2024.102769] [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: 02/27/2024] [Revised: 05/18/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
Many strategies for regenerating the damaged tissues or degenerating cells are employed in regenerative medicine. Stem cell technology is a modern strategy of the recent approaches, particularly the use of mesenchymal stem cells (MCSs). The ability of MSCs to differentiate as well as their characteristic behaviour as paracrine effector has established them as key elements in tissue repair. Recently, extracellular vesicles (EVs) shed by MSCs have emerged as a promising cell free therapy. This comprehensive review encompasses MSCs-derived exosomes and their therapeutic potential as nanotherapeutics. We also discuss their potency as drug delivery nano-carriers in comparison with liposomes. A better knowledge of EVs behaviour in vivo and of their mechanism of action are key to determine parameters of an optimal formulation in pilot studies and to establish industrial processes.
Collapse
Affiliation(s)
- Ali Imran Abid
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France
| | - Guillaume Conzatti
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France.
| | - Florence Toti
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Nicolas Anton
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Thierry Vandamme
- UMR 1260, Regenerative Nanomedicine (RNM), INSERM (French National Institute of Health and Medical Research), University of Strasbourg, F-67000 Strasbourg, France; Faculty of Pharmacy, University of Strasbourg, 67400 Illkirch-Graffenstaden, France.
| |
Collapse
|
9
|
Li Y, Wang Y, Zhang Y, Zhu Y, Dong Y, Cheng H, Zhang Y, Wang Y, Li Z, Gao J. Engineered mesenchymal stem cell-derived extracellular vesicles: kill tumors and protect organs. Theranostics 2024; 14:6202-6217. [PMID: 39431009 PMCID: PMC11488101 DOI: 10.7150/thno.99618] [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: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 10/22/2024] Open
Abstract
Solid tumors cause 90% of cancers and remain the primary cause of mortality. However, treating solid tumors presents significant challenges due to the complex tumor microenvironment and drug resistance, leading to inadequate treatment targeting and severe side effects. Surgery, radiotherapy, and chemotherapy Although it is an effective method for the treatment of solid tumors, it can lead to organ dysfunction and affect patient prognosis. Therefore, it is imperative to improve treatment precision and organ repair capabilities to manage solid tumors. Mesenchymal stem cell extracellular vesicles (MSC-EVs) have wide application prospects as a new agent for solid tumor therapy. Firstly, MSC-EVs is a derivative of MSCs. It has the function of promoting tissue regeneration by inducing dedifferentiation in surviving cells after injury. Additionally, MSC-EVs offer unique advantages in terms of safety, stability and penetrability, making them a promising extracellular therapeutic modality for solid tumor treatment. Finally, MSC-EVs are able to enhance therapeutic efficacy through engineering strategies. To sum up, this review takes MSC-EVs as its object. And then we discuss recent advancements and engineering strategies in the use of MSC-EVs for soid tumor suppression. This review aims to inspire researchers to devise a new method for effectively treat solid tumors.
Collapse
Affiliation(s)
- Yu Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Stem Cell and Regeneration Medicine Institute, Research Center of Translational Medicine, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yao Wang
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Heilongjiang Mudanjiang, 157011, China
| | - Yu Zhang
- Shanghai Key Laboratory of Cell Engineering, Shanghai, 200120, China
| | - Yuruchen Zhu
- School of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yuhui Dong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Haobin Cheng
- School of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Yue Wang
- Stem Cell and Regeneration Medicine Institute, Research Center of Translational Medicine, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, 200120, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- National Key Laboratory of lmmunology and Inflammation, Naval Medical University, Shanghai, 200433, China
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Jie Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Changhai Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| |
Collapse
|
10
|
Singer J, Knezic N, Layne J, Gohring G, Christiansen J, Rothrauff B, Huard J. Enhancing Cartilage Repair: Surgical Approaches, Orthobiologics, and the Promise of Exosomes. Life (Basel) 2024; 14:1149. [PMID: 39337932 PMCID: PMC11432843 DOI: 10.3390/life14091149] [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: 07/28/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Treating cartilage damage is challenging as its ability for self-regeneration is limited. Left untreated, it can progress to osteoarthritis (OA), a joint disorder characterized by the deterioration of articular cartilage and other joint tissues. Surgical options, such as microfracture and cell/tissue transplantation, have shown promise as techniques to harness the body's endogenous regenerative capabilities to promote cartilage repair. Nonetheless, these techniques have been scrutinized due to reported inconsistencies in long-term outcomes and the tendency for the defects to regenerate as fibrocartilage instead of the smooth hyaline cartilage native to joint surfaces. Orthobiologics are medical therapies that utilize biologically derived substances to augment musculoskeletal healing. These treatments are rising in popularity because of their potential to enhance surgical standards of care. More recent developments in orthobiologics have focused on the role of exosomes in articular cartilage repair. Exosomes are nano-sized extracellular vesicles containing cargo such as proteins, lipids, and nucleic acids, and are known to facilitate intercellular communication, though their regenerative potential still needs to be fully understood. This review aims to demonstrate the advancements in cartilage regeneration, highlight surgical and biological treatment options, and discuss the recent strides in understanding the precise mechanisms of action involved.
Collapse
Affiliation(s)
- Jacob Singer
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Noah Knezic
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jonathan Layne
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Greta Gohring
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jeff Christiansen
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Ben Rothrauff
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Johnny Huard
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
11
|
Wu J, Wu J, Liu Z, Gong Y, Feng D, Xiang W, Fang S, Chen R, Wu Y, Huang S, Zhou Y, Liu N, Xu H, Zhou S, Liu B, Ni Z. Mesenchymal stem cell-derived extracellular vesicles in joint diseases: Therapeutic effects and underlying mechanisms. J Orthop Translat 2024; 48:53-69. [PMID: 39170747 PMCID: PMC11338158 DOI: 10.1016/j.jot.2024.07.005] [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: 03/05/2024] [Revised: 06/09/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Joint diseases greatly impact the daily lives and occupational functioning of patients globally. However, conventional treatments for joint diseases have several limitations, such as unsatisfatory efficacy and side effects, necessitating the exploration of more efficacious therapeutic strategies. Mesenchymal stem cell (MSC)-derived EVs (MSC-EVs) have demonstrated high therapeutic efficacyin tissue repair and regeneration, with low immunogenicity and tumorigenicity. Recent studies have reported that EVs-based therapy has considerable therapeutic effects against joint diseases, including osteoarthritis, tendon and ligament injuries, femoral head osteonecrosis, and rheumatoid arthritis. Herein, we review the therapeutic potential of various types of MSC-EVs in the aforementioned joint diseases, summarise the mechanisms underlying specific biological effects of MSC-EVs, and discuss future prospects for basic research on MSC-EV-based therapeutic modalities and their clinical translation. In general, this review provides an in-depth understanding of the therapeutic effects of MSC-EVs in joint diseases, as well as the underlying mechanisms, which may be beneficial to the clinical translation of MSC-EV-based treatment. The translational potential of this article: MSC-EV-based cell-free therapy can effectively promote regeneration and tissue repair. When used to treat joint diseases, MSC-EVs have demonstrated desirable therapeutic effects in preclinical research. This review may supplement further research on MSC-EV-based treatment of joint diseases and its clinical translation.
Collapse
Affiliation(s)
- Jinhui Wu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Jiangyi Wu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Zheng Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yunquan Gong
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Daibo Feng
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Wei Xiang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Shunzheng Fang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, China
| | - Yaran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing, 400038, China
| | - Shu Huang
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yizhao Zhou
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Ningning Liu
- Department of Laboratory Medicine, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, 450003, China
| | - Hao Xu
- Department of Laboratory Medicine, the Third Affiliated Hospital of Zhengzhou University Zhengzhou, 450003, China
| | - Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, China
| | - Baorong Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Zhenhong Ni
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| |
Collapse
|
12
|
Zeng B, Li Y, Khan N, Su A, Yang Y, Mi P, Jiang B, Liang Y, Duan L. Yin-Yang: two sides of extracellular vesicles in inflammatory diseases. J Nanobiotechnology 2024; 22:514. [PMID: 39192300 PMCID: PMC11351009 DOI: 10.1186/s12951-024-02779-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
The concept of Yin-Yang, originating in ancient Chinese philosophy, symbolizes two opposing but complementary forces or principles found in all aspects of life. This concept can be quite fitting in the context of extracellular vehicles (EVs) and inflammatory diseases. Over the past decades, numerous studies have revealed that EVs can exhibit dual sides, acting as both pro- and anti-inflammatory agents, akin to the concept of Yin-Yang theory (i.e., two sides of a coin). This has enabled EVs to serve as potential indicators of pathogenesis or be manipulated for therapeutic purposes by influencing immune and inflammatory pathways. This review delves into the recent advances in understanding the Yin-Yang sides of EVs and their regulation in specific inflammatory diseases. We shed light on the current prospects of engineering EVs for treating inflammatory conditions. The Yin-Yang principle of EVs bestows upon them great potential as, therapeutic, and preventive agents for inflammatory diseases.
Collapse
Affiliation(s)
- Bin Zeng
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
- Graduate School, Guangxi University of Chinese Medicine, Nanning, 53020, Guangxi, China
| | - Ying Li
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Nawaz Khan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Aiyuan Su
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Yicheng Yang
- Eureka Biotech Inc, Philadelphia, PA, 19104, USA
| | - Peng Mi
- Department of Radiology, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bin Jiang
- Eureka Biotech Inc, Philadelphia, PA, 19104, USA.
| | - Yujie Liang
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China.
| | - Li Duan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China.
| |
Collapse
|
13
|
Vadhan A, Gupta T, Hsu WL. Mesenchymal Stem Cell-Derived Exosomes as a Treatment Option for Osteoarthritis. Int J Mol Sci 2024; 25:9149. [PMID: 39273098 PMCID: PMC11395657 DOI: 10.3390/ijms25179149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Osteoarthritis (OA) is a leading cause of pain and disability worldwide in elderly people. There is a critical need to develop novel therapeutic strategies that can effectively manage pain and disability to improve the quality of life for older people. Mesenchymal stem cells (MSCs) have emerged as a promising cell-based therapy for age-related disorders due to their multilineage differentiation and strong paracrine effects. Notably, MSC-derived exosomes (MSC-Exos) have gained significant attention because they can recapitulate MSCs into therapeutic benefits without causing any associated risks compared with direct cell transplantation. These exosomes help in the transport of bioactive molecules such as proteins, lipids, and nucleic acids, which can influence various cellular processes related to tissue repair, regeneration, and immune regulation. In this review, we have provided an overview of MSC-Exos as a considerable treatment option for osteoarthritis. This review will go over the underlying mechanisms by which MSC-Exos may alleviate the pathological hallmarks of OA, such as cartilage degradation, synovial inflammation, and subchondral bone changes. Furthermore, we have summarized the current preclinical evidence and highlighted promising results from in vitro and in vivo studies, as well as progress in clinical trials using MSC-Exos to treat OA.
Collapse
Affiliation(s)
- Anupama Vadhan
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin 632007, Taiwan;
| | - Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan;
| | - Wen-Li Hsu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin 632007, Taiwan;
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| |
Collapse
|
14
|
Chaibakhsh S, Azimi F, Shoae-Hassani A, Niknam P, Ghamari A, Dehghan S, Nilforushan N. Evaluating the impact of mesenchymal stem cell therapy on visual acuity and retinal nerve fiber layer thickness in optic neuropathy patients: a comprehensive systematic review and meta-analysis. BMC Ophthalmol 2024; 24:316. [PMID: 39075477 PMCID: PMC11287858 DOI: 10.1186/s12886-024-03588-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Stem cell therapy has emerged as a potential therapeutic avenue for optic neuropathy patients. To assess its safety and efficacy, we conducted a systematic review and meta-analysis, focusing on the latest evidence pertaining to the improvement of visual acuity (VA) through stem cell therapy. METHODS We analyzed Each database from its inception until June 2024. PubMed, Scopus, and Google Scholar were systematically searched to identify the included studies. Data were extracted regarding the year of publication, the name of the first author, sample size, VA (Log Mar), and Retinal Nerve Fiber Layer (RNFL) thickness. PRISMA protocol was used as a guide to perform this meta-analysis. STATA 16 was used for statistical analysis. RESULTS A total of 66 eyes were examined in seven papers. Based on the meta-analysis, the mean VA (Log MAR) of patients with optic neuropathy improved from 0.90 to 0.65 following stem cell therapy intervention (p-value = 0.001). The thickness of the RNFLs did not demonstrate a significant change (p-value was 0.174). CONCLUSION According to this systematic review and meta-analysis, stem cell therapy may improve the visual acuity of patients with optic neuropathy. Aside from the traditional therapy that can be provided to patients with optic neuropathy, stem cell therapy may also be beneficial.
Collapse
Affiliation(s)
- Samira Chaibakhsh
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Azimi
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Shoae-Hassani
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
- Stem Cells and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Parvin Niknam
- Department of Ophthalmology, Mayo clinic, Rochester, MN, USA
| | - Ali Ghamari
- Pediatric Cell and Gene Therapy Research Center, Cell & Tissue Research Institute, Tehran university of Medical Sciences, Gene, Tehran, Iran
| | - Samaneh Dehghan
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.
- Stem Cells and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Naveed Nilforushan
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Mei R, Wan Z, Yang C, Shen X, Wang R, Zhang H, Yang R, Li J, Song Y, Su H. Advances and clinical challenges of mesenchymal stem cell therapy. Front Immunol 2024; 15:1421854. [PMID: 39100671 PMCID: PMC11294097 DOI: 10.3389/fimmu.2024.1421854] [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: 04/23/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024] Open
Abstract
In recent years, cell therapy has provided desirable properties for promising new drugs. Mesenchymal stem cells are promising candidates for developing genetic engineering and drug delivery strategies due to their inherent properties, including immune regulation, homing ability and tumor tropism. The therapeutic potential of mesenchymal stem cells is being investigated for cancer therapy, inflammatory and fibrotic diseases, among others. Mesenchymal stem cells are attractive cellular carriers for synthetic nanoparticles for drug delivery due to their inherent homing ability. In this review, we comprehensively discuss the various genetic and non-genetic strategies of mesenchymal stem cells and their derivatives in drug delivery, tumor therapy, immune regulation, tissue regeneration and other fields. In addition, we discuss the current limitations of stem cell therapy and the challenges in clinical translation, aiming to identify important development areas and potential future directions.
Collapse
Affiliation(s)
- Ruiyan Mei
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Zhuo Wan
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Cheng Yang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Xiangjing Shen
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Ronglin Wang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haihua Zhang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Rui Yang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Junqiang Li
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Yang Song
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
16
|
Li L, Zeng L, Wu W. Study on the mechanism of quercetin inducing mesenchymal stem cells to differentiate into fibroblasts through TGF-β1 and IGF-1. Tissue Cell 2024; 88:102383. [PMID: 38613933 DOI: 10.1016/j.tice.2024.102383] [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: 02/20/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Stem cell differentiation has opened up new avenues for disease treatment, tissue repair, and drug development in the study of regenerative medicine, and has huge application prospects. This study aimed to explore the mechanism of quercetin on the differentiation of mesenchymal stem cells (MSCs) into fibroblasts. METHODS In this study, cell differentiation experiments and flow cytometry were used to detect the successful isolation of bone marrow MSCs from SD rats. Quercetin at 5, 10, and 20 μM was used as low, medium, and high doses to intervene in MSCs. The cell viability changes of ligament fibroblasts at 24, 48, and 72 hours after quercetin treatment were detected using a CCK-8 cell counting kit. Cell proliferative capacity was determined by flow cytometry. RT-qPCR measured the relative expression levels of TGF-β1, IGF-1, COL-Ⅰ, COL-Ⅲ, FN (fibronectin), and TNMD (Tenomodulin) in different experimental groups. Molecular docking experiments were conducted to explore the binding effect of quercetin on TGF-β1 and IGF-1 proteins. RESULTS Flow cytometry verified the successful isolation of MSCs, which had high expression of CD29 and CD73, while lower expression of CD90 and CD45. Experimental results show that low and medium doses of quercetin can enhance cell proliferation, while high doses have no significant effect on cells. Detection of cell proliferation through flow cytometry yielded similar results to CCK-8. Transwell experiments have shown that low and medium doses of quercetin can increase cell migration ability. In addition, RT-qPCR detection showed that quercetin can increase the mRNA expression of TGF-β1 and IGF-1, and promote the expression of COL-Ⅰ, COL-Ⅲ, FN, and TNMD genes in ligament fibroblasts. Molecular docking results showed that quercetin can bind firmly to TGF-β1 and IGF-1. CONCLUSION Overall, this study revealed the morphological characteristics and identification of MSCs, as well as the regulatory mechanism of quercetin on the behavior of ligament fibroblasts. Quercetin affects the proliferation and gene expression of ligament fibroblasts by regulating the expression of TGF-β1 and IGF-1, which may provide a new perspective for biomedical research on the skeletal system.
Collapse
Affiliation(s)
- Liji Li
- Liwan District Orthopedics Hospital Rehabilitation Department, China.
| | - Liang Zeng
- Liwan District Orthopedics Hospital Rehabilitation Department, China
| | - Weizhi Wu
- Liwan District Orthopedics Hospital Rehabilitation Department, China.
| |
Collapse
|
17
|
Liao HJ, Yang YP, Liu YH, Tseng HC, Huo TI, Chiou SH, Chang CH. Harnessing the potential of mesenchymal stem cells-derived exosomes in degenerative diseases. Regen Ther 2024; 26:599-610. [PMID: 39253597 PMCID: PMC11382214 DOI: 10.1016/j.reth.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have gained attention as a promising therapeutic approach in both preclinical and clinical osteoarthritis (OA) settings. Various joint cell types, such as chondrocytes, synovial fibroblasts, osteoblasts, and tenocytes, can produce and release extracellular vesicles (EVs), which subsequently influence the biological activities of recipient cells. Recently, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have shown the potential to modulate various physiological and pathological processes through the modulation of cellular differentiation, immune responses, and tissue repair. This review explores the roles and therapeutic potential of MSC-EVs in OA and rheumatoid arthritis, cardiovascular disease, age-related macular degeneration, Alzheimer's disease, and other degenerative diseases. Notably, we provide a comprehensive summary of exosome biogenesis, microRNA composition, mechanisms of intercellular transfer, and their evolving role in the highlight of exosome-based treatments in both preclinical and clinical avenues.
Collapse
Affiliation(s)
- Hsiu-Jung Liao
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Hao Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Huan-Chin Tseng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Teh-Ia Huo
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Hung Chang
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City, Taiwan
| |
Collapse
|
18
|
Aouabdi S, Nedjadi T, Alsiary R, Mouffouk F, Ansari HR. Transcriptomics Demonstrates Significant Biological Effect of Growing Stem Cells on RGD-Cotton Scaffold. Tissue Eng Part A 2024. [PMID: 38666698 DOI: 10.1089/ten.tea.2023.0333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024] Open
Abstract
Stem cell therapy provides a viable alternative treatment for degenerated or damaged tissue. Stem cells have been used either alone or in conjunction with an artificial scaffold. The latter provides a structural advantage by enabling the cells to thrive in three-dimensional (3D) settings, closely resembling the natural in vivo environments. Previously, we disclosed the development of a 3D scaffold made from cotton, which was conjugated with arginyl-glycyl-aspartic acid (RGD), to facilitate the growth and proliferation of mesenchymal stem cells (MSCs). This scaffold allowed the MSCs to adhere and proliferate without compromising their viability or their stem cell markers. A comprehensive analysis investigation of the molecular changes occurring in MSCs adhering to the cotton fibers will contribute to the advancement of therapy. The objective of this study is to analyze the molecular processes occurring in the growth of MSCs on a cotton-RGD conjugated-based scaffold by examining their gene expression profiles. To achieve this, we conducted an experiment where MSCs were seeded with and without the scaffold for a duration of 48 h. Subsequently, cells were collected for RNA extraction, cDNA synthesis, and whole-transcriptomic analysis performed on both populations. Our analysis revealed several upregulated and downregulated differently expressed genes in the MSCs adhering to the scaffold compared with the control cells. Through gene ontology analysis, we were able to identify enriched biological processes, molecular functions, pathways, and protein-protein interactions in these differentially expressed genes. Our data suggest that the scaffold may have the potential to enhance osteogenesis in the MSCs. Furthermore, our results indicate that the scaffold does not induce oxidative stress, inflammation, or aging in the MSCs. These findings provide valuable insights for the application of MSCs in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Sihem Aouabdi
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Taoufik Nedjadi
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Rawiah Alsiary
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Fouzi Mouffouk
- Department of Chemistry, Kuwait University, Kuwait, Kuwait
| | - Hifzur Rahman Ansari
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| |
Collapse
|
19
|
Wang J, Ye W, Jiang M, Zhou Y, Zheng J. Therapeutic potential of exosome derived from hepatocyte growth factor-overexpressing adipose mesenchymal stem cells in TGFβ1-stimulated hepatic stellate cells. Cytotechnology 2024; 76:217-229. [PMID: 38495297 PMCID: PMC10940570 DOI: 10.1007/s10616-023-00611-0] [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: 07/18/2023] [Accepted: 12/14/2023] [Indexed: 03/19/2024] Open
Abstract
Cirrhosis is a familiar end-stage of multiple chronic liver diseases. The gene-modified mesenchymal stem cells (MSCs) have become one of the most promising schemes for the treatment of cirrhosis. MSCs exhibit their therapeutic role mainly by secreting hepatocyte growth factor (HGF). The aim of this research was to probe the anti-fibrosis role of exosomes secreted by HGF modified-mouse adipose MSCs (ADMSCs) on activated hepatic stellate cells (HSCs) and to preliminarily explore the possible mechanism. Firstly, mouse ADMSCs were isolated and identified. Quantitative real-time polymerase chain reaction verified the transfection efficiency of ADMSC transfected with HGF lentivirus. Exosomes derived from ADMSC transfecting negative control/HGF (ADMSCNC-Exo/ADMSCHGF-Exo) were extracted by density gradient centrifugation. HSCs were allocated to the control, TGF-β, TGF-β + ADMSC-Exo, TGF-β + ADMSCNC-Exo, and TGF-β + ADMSCHGF-Exo groups. Moreover, all mice were distributed to the control, CCl4 (40% CCl4 in olive oil), CCl4+ADMSC-Exo, CCl4+ADMSCNC-Exo, and CCl4+ADMSCHGF-Exo groups. Exosomes derived from ADMSCs with or without HGF transfection suppressed HSC activation, as evidenced by attenuating cell viability and cell cycle arrest at S phase but inducing apoptosis. Moreover, ADMSC-Exo, ADMSCNC-Exo, and ADMSCHGF-Exo effectively repressed the gene and protein levels of α-SMA, Col-I, Rho A, Cdc42, and Rac1 in TGF-β-treated HSCs, and ADMSCHGF-Exo had the best effect. ADMSCHGF-Exo had a stronger regulatory effect on serum liver index than ADMSCNC-Exo in CCl4-induced mice. In conclusion, ADMSCHGF-Exo alleviated liver fibrosis by weakening the Rho pathway, thus reducing collagen production.
Collapse
Affiliation(s)
- Jin Wang
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Weikang Ye
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Ming Jiang
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Yinong Zhou
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Jie Zheng
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| |
Collapse
|
20
|
Wen S, Huang X, Ma J, Zhao G, Ma T, Chen K, Huang G, Chen J, Shi J, Wang S. Exosomes derived from MSC as drug system in osteoarthritis therapy. Front Bioeng Biotechnol 2024; 12:1331218. [PMID: 38576449 PMCID: PMC10993706 DOI: 10.3389/fbioe.2024.1331218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/08/2024] [Indexed: 04/06/2024] Open
Abstract
Osteoarthritis (OA) is the most common degenerative disease of the joint with irreversible cartilage damage as the main pathological feature. With the development of regenerative medicine, mesenchymal stem cells (MSCs) have been found to have strong therapeutic potential. However, intraarticular MSCs injection therapy is limited by economic costs and ethics. Exosomes derived from MSC (MSC-Exos), as the important intercellular communication mode of MSCs, contain nucleic acid, proteins, lipids, microRNAs, and other biologically active substances. With excellent editability and specificity, MSC-Exos function as a targeted delivery system for OA treatment, modulating immunity, inhibiting apoptosis, and promoting regeneration. This article reviews the mechanism of action of MSC-Exos in the treatment of osteoarthritis, the current research status of the preparation of MSC-Exos and its application of drug delivery in OA therapy.
Collapse
Affiliation(s)
- Shuzhan Wen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xin Huang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingchun Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Guanglei Zhao
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Tiancong Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Kangming Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Gangyong Huang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingsheng Shi
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Siqun Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [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: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. 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 developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
22
|
Luo D, Zhu H, Li S, Wang Z, Xiao J. Mesenchymal stem cell-derived exosomes as a promising cell-free therapy for knee osteoarthritis. Front Bioeng Biotechnol 2024; 12:1309946. [PMID: 38292826 PMCID: PMC10824863 DOI: 10.3389/fbioe.2024.1309946] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Osteoarthritis (OA), as a degenerative disease, leads to high socioeconomic burdens and disability rates. The knee joint is typically the most affected and is characterized by progressive destruction of articular cartilage, subchondral bone remodeling, osteophyte formation and synovial inflammation. The current management of OA mainly focuses on symptomatic relief and does not help to slow down the advancement of disease. Recently, mesenchymal stem cells (MSCs) and their exosomes have garnered significant attention in regenerative therapy and tissue engineering areas. Preclinical studies have demonstrated that MSC-derived exosomes (MSC-Exos), as bioactive factor carriers, have promising results in cell-free therapy of OA. This study reviewed the application of various MSC-Exos for the OA treatment, along with exploring the potential underlying mechanisms. Moreover, current strategies and future perspectives for the utilization of engineered MSC-Exos, alongside their associated challenges, were also discussed.
Collapse
Affiliation(s)
| | | | | | - Zhenggang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Tan F, Li X, Wang Z, Li J, Shahzad K, Zheng J. Clinical applications of stem cell-derived exosomes. Signal Transduct Target Ther 2024; 9:17. [PMID: 38212307 PMCID: PMC10784577 DOI: 10.1038/s41392-023-01704-0] [Citation(s) in RCA: 178] [Impact Index Per Article: 178.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/15/2023] [Accepted: 11/12/2023] [Indexed: 01/13/2024] Open
Abstract
Although stem cell-based therapy has demonstrated considerable potential to manage certain diseases more successfully than conventional surgery, it nevertheless comes with inescapable drawbacks that might limit its clinical translation. Compared to stem cells, stem cell-derived exosomes possess numerous advantages, such as non-immunogenicity, non-infusion toxicity, easy access, effortless preservation, and freedom from tumorigenic potential and ethical issues. Exosomes can inherit similar therapeutic effects from their parental cells such as embryonic stem cells and adult stem cells through vertical delivery of their pluripotency or multipotency. After a thorough search and meticulous dissection of relevant literature from the last five years, we present this comprehensive, up-to-date, specialty-specific and disease-oriented review to highlight the surgical application and potential of stem cell-derived exosomes. Exosomes derived from stem cells (e.g., embryonic, induced pluripotent, hematopoietic, mesenchymal, neural, and endothelial stem cells) are capable of treating numerous diseases encountered in orthopedic surgery, neurosurgery, plastic surgery, general surgery, cardiothoracic surgery, urology, head and neck surgery, ophthalmology, and obstetrics and gynecology. The diverse therapeutic effects of stem cells-derived exosomes are a hierarchical translation through tissue-specific responses, and cell-specific molecular signaling pathways. In this review, we highlight stem cell-derived exosomes as a viable and potent alternative to stem cell-based therapy in managing various surgical conditions. We recommend that future research combines wisdoms from surgeons, nanomedicine practitioners, and stem cell researchers in this relevant and intriguing research area.
Collapse
Affiliation(s)
- Fei Tan
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China.
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China.
- The Royal College of Surgeons in Ireland, Dublin, Ireland.
- The Royal College of Surgeons of England, London, UK.
| | - Xuran Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Zhao Wang
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
| | - Jiaojiao Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Khawar Shahzad
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
- Plasma Medicine and Surgical Implants Center, Tongji University, Shanghai, China
| | - Jialin Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University, Shanghai, China
| |
Collapse
|
24
|
Yuan J, Wang Y, Xu X, Yang M, Fan Y, Shi X, Sun L, Shan M, Ma L. Effects of Different Light Environments with Varying Spectral Composition on the Axial Lengths and Scleral Specificity Protein 1 and Collagen Type I Expression in Juvenile Guinea Pigs. Folia Biol (Praha) 2024; 70:219-228. [PMID: 39692576 DOI: 10.14712/fb2024070040219] [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] [Indexed: 12/19/2024]
Abstract
The study aimed to investigate changes in the eye axial length in juvenile guinea pigs and the expression of scleral specificity protein 1 (Sp1) and collagen type I (Col-I) under different light environments with varying spectral composition. The animals were randomly divided into five groups: natural light (N), LED light with a low colour temperature (L), E light (E), Fulia light (F), and Gulia light (G). Axial lengths were measured every two weeks, and the expression of Sp1 and Col-I in the sclera was assessed by immunohistochemistry, Western blot and RT-qPCR. After 4, 6, 8, 10, and 12 weeks of light exposure, the L and G groups showed considerably longer axial lengths than the N group, with the L group exhibiting significantly longer axial lengths compared with the E and F groups. The protein and mRNA expression levels of Sp1 and Col-I, ranked from highest to lowest, were as follows: N, E, F, G, and L. The expression of Sp1 and Col-I was positively correlated, but both were negatively correlated with the length of the eye axis. The E group demonstrated higher Sp1 and Col-I expression than the other artificial light groups. Artificial light with a continuous, full spectrum lacking peaks and valleys can inhibit the elongation of the eye axis in juvenile guinea pigs and has a protective effect against myopia. There may be a certain relationship between Sp1 and Col-I, and the transforming growth factor-β1-Sp1-Col-I signalling pathway may play a crucial role in myopic scleral extracellular matrix remodelling.
Collapse
Affiliation(s)
- Jianbao Yuan
- Department of Ophthalmology, Clinical College of Yizheng People's Hospital, Jiangsu Health Vocational College, Yangzhou, Jiangsu, China.
| | - Yuliang Wang
- Department of Ophthalmology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xinyu Xu
- Department of Ophthalmology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mei Yang
- Department of Ophthalmology, Clinical College of Yizheng People's Hospital, Jiangsu Health Vocational College, Yangzhou, Jiangsu, China
| | - Yipeng Fan
- Department of Ophthalmology, Clinical College of Yizheng People's Hospital, Jiangsu Health Vocational College, Yangzhou, Jiangsu, China
| | - Xiaopan Shi
- Department of Ophthalmology, Clinical College of Yizheng People's Hospital, Jiangsu Health Vocational College, Yangzhou, Jiangsu, China
| | - Lulu Sun
- Department of Ophthalmology, Clinical College of Yizheng People's Hospital, Jiangsu Health Vocational College, Yangzhou, Jiangsu, China
| | - Mingyu Shan
- Department of Ophthalmology, Clinical College of Yizheng People's Hospital, Jiangsu Health Vocational College, Yangzhou, Jiangsu, China
| | - Lei Ma
- Department of Ophthalmology, Clinical College of Yizheng People's Hospital, Jiangsu Health Vocational College, Yangzhou, Jiangsu, China
| |
Collapse
|
25
|
Xie W, Luo T, Ma Z, Xue S, Jia X, Yang T, Song Z. Tumor Necrosis Factor Alpha Preconditioned Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Enhance the Inhibition of Necroptosis of Acinar cells in Severe Acute Pancreatitis. Tissue Eng Part A 2023; 29:607-619. [PMID: 37565286 DOI: 10.1089/ten.tea.2023.0139] [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] [Indexed: 08/12/2023] Open
Abstract
Severe acute pancreatitis (SAP) is a common abdominal emergency with a high mortality rate and a lack of effective therapeutic options. Although mesenchymal stem cell (MSC) transplantation is a potential treatment for SAP, the mechanism remains unclear. It has been suggested that MSCs may act mainly through paracrine effects; therefore, we aimed to demonstrate the therapeutic efficacy of extracellular vesicles (EVs) derived from human umbilical cord mesenchymal stem cells (UCMSCs) for SAP. Na-taurocholate was used to induce a rat SAP model through retrograde injection into the common biliopancreatic duct. After 72 h of EVs transplantation, pancreatic pathological damage was alleviated, along with a decrease in serum amylase activity and pro-inflammatory cytokine levels. Interestingly, when UCMSCs were preconditioned with 10 ng/mL tumor necrosis factor alpha (TNF-α) for 48 h, the obtained EVs (named TNF-α-EVs) performed an enhanced efficacy. Furthermore, both animal and cellular experiments showed that TNF-α-EVs alleviated the necroptosis of acinar cells of SAP through RIPK3/MLKL axis. In conclusion, our study demonstrated that TNF-α-EVs were able to enhance the therapeutic effect on SAP by inhibiting necroptosis compared to normal EVs. This study heralds that TNF-α-EVs may be a promising therapeutic approach for SAP in the future.
Collapse
Affiliation(s)
- Wangcheng Xie
- Department of General Surgery and Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Hepatic-Biliary-Pancreatic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingyi Luo
- Department of General Surgery and Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Hepatic-Biliary-Pancreatic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhilong Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaobo Xue
- Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuyang Jia
- Department of Metabolic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingsong Yang
- Department of General Surgery and Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenshun Song
- Department of Hepatic-Biliary-Pancreatic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
26
|
Guo R, Fan J. Extracellular Vesicles Derived from Auricular Chondrocytes Facilitate Cartilage Differentiation of Adipose-Derived Mesenchymal Stem Cells. Aesthetic Plast Surg 2023; 47:2823-2832. [PMID: 36849663 DOI: 10.1007/s00266-023-03292-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/20/2023] [Indexed: 03/01/2023]
Abstract
PURPOSE Adipose-derived mesenchymal stem cell (ADSC)-based therapies have been utilized for cartilage regeneration because of their multi-lineage differentiation ability. However, commonly used cartilage inducers such as the transforming growth factor beta-3 (TGF-β3) may be prone to cartilage dedifferentiation and hypertrophy. The directional differentiation of elastic cartilage is limited nowadays. Extracellular vesicles (EVs) have been reported to influence the specific differentiation of mesenchymal stem cells (MSCs) by reflecting the composition of the parental cells. However, the role of auricular chondrogenic-derived EVs (AC-EVs) in elastic chondrogenic differentiation of ADSCs has not yet been reported. RESULTS AC-EVs isolated from the external ears of swine exhibited a positive effect on cell proliferation and migration. Furthermore, AC-EVs efficiently promoted chondrogenic differentiation of ADSCs in pellet culture, as shown by the elevated levels of COL2A1, ACAN, and SOX-9 expression. Moreover, there was a significantly higher expression of elastin and a lower expression of the fibrotic marker COL1A1 in comparison with that achieved with TGF-β3. The staining results demonstrated that AC-EVs promoted the deposition of cartilage-specific matrix, which is in good concordance with the real-time polymerase chain reaction (RT-PCR) results. CONCLUSIONS Auricular chondrogenic-derived EVs are a crucial component in elastic chondrogenic differentiation and other biological behaviors of ADSCs, which may be a useful ingredient for cartilage tissue engineering and external ear reconstruction. NO LEVEL ASSIGNED This journal requires that authors 42 assign a level of evidence to each submission to which 43 Evidence-Based Medicine rankings are applicable. This 44 excludes Review Articles, Book Reviews, and manuscripts 45 that concern Basic Science, Animal Studies, Cadaver 46 Studies, and Experimental Studies. For a full description of 47 these Evidence-Based Medicine ratings, please refer to the 48 Table oôf Contents or the online Instructions to Authors 49 www.springer.com/00266 .
Collapse
Affiliation(s)
- Rui Guo
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Jincai Fan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| |
Collapse
|
27
|
Lv S, Wang G, Dai L, Wang T, Wang F. Cellular and Molecular Connections Between Bone Fracture Healing and Exosomes. Physiol Res 2023; 72:565-574. [PMID: 38015756 PMCID: PMC10751053 DOI: 10.33549/physiolres.935143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/25/2023] [Indexed: 01/05/2024] Open
Abstract
Fracture healing is a multifaceted process that requires various phases and intercellular interactions. In recent years, investigations have been conducted to assess the feasibility of utilizing exosomes, small extracellular vesicles (EVs), to enhance and accelerate the healing process. Exosomes serve as a cargo transport platform, facilitating intercellular communication, promoting the presentation of antigens to dendritic cells, and stimulating angiogenesis. Exosomes have a special structure that gives them a special function, especially in the healing process of bone injuries. This article provides an overview of cellular and molecular processes associated with bone fracture healing, as well as a survey of existing exosome research in this context. We also discuss the potential use of exosomes in fracture healing, as well as the obstacles that must be overcome to make this a viable clinical practice.
Collapse
Affiliation(s)
- S Lv
- Department of Orthopedics, Sinopharm China Railway Engineering Corporation Central Hospital, Hefei, China.
| | | | | | | | | |
Collapse
|
28
|
Li Z, Bi R, Zhu S. The Dual Role of Small Extracellular Vesicles in Joint Osteoarthritis: Their Global and Non-Coding Regulatory RNA Molecule-Based Pathogenic and Therapeutic Effects. Biomolecules 2023; 13:1606. [PMID: 38002288 PMCID: PMC10669328 DOI: 10.3390/biom13111606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
OA is the most common joint disease that affects approximately 7% of the global population. Current treatment methods mainly relieve its symptoms with limited repairing effect on joint destructions, which ultimately contributes to the high morbidity rate of OA. Stem cell treatment is a potential regenerative medical therapy for joint repair in OA, but the uncertainty in differentiation direction and immunogenicity limits its clinical usage. Small extracellular vesicles (sEVs), the by-products secreted by stem cells, show similar efficacy levels but have safer regenerative repair effect without potential adverse outcomes, and have recently drawn attention from the broader research community. A series of research works and reviews have been performed in the last decade, providing references for the application of various exogenous therapeutic sEVs for treating OA. However, the clinical potential of target intervention involving endogenous pathogenic sEVs in the treatment of OA is still under-explored and under-discussed. In this review, and for the first time, we emphasize the dual role of sEVs in OA and explain the effects of sEVs on various joint tissues from both the pathogenic and therapeutic aspects. Our aim is to provide a reference for future research in the field.
Collapse
Affiliation(s)
- Zhi Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Ruiye Bi
- Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Songsong Zhu
- Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
29
|
Huang S, Liu Y, Wang C, Xiang W, Wang N, Peng L, Jiang X, Zhang X, Fu Z. Strategies for Cartilage Repair in Osteoarthritis Based on Diverse Mesenchymal Stem Cells-Derived Extracellular Vesicles. Orthop Surg 2023; 15:2749-2765. [PMID: 37620876 PMCID: PMC10622303 DOI: 10.1111/os.13848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Osteoarthritis (OA) causes disability and significant economic and social burden. Cartilage injury is one of the main pathological features of OA, and is often manifested by excessive chondrocyte death, inflammatory response, abnormal bone metabolism, imbalance of extracellular matrix (ECM) metabolism, and abnormal vascular or nerve growth. Regrettably, due to the avascular nature of cartilage, its capacity to repair is notably limited. Mesenchymal stem cells-derived extracellular vesicles (MSCs-EVs) play a pivotal role in intercellular communication, presenting promising potential not only as early diagnostic biomarkers in OA but also as efficacious therapeutic strategy. MSCs-EVs were confirmed to play a therapeutic role in the pathological process of cartilage injury mentioned above. This paper comprehensively provides the functions and mechanisms of MSCs-EVs in cartilage repair.
Collapse
Affiliation(s)
- Shanjun Huang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Yujiao Liu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Chenglong Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Wei Xiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Nianwu Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Li Peng
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xuanang Jiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiaomin Zhang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Zhijiang Fu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
30
|
Bertolino GM, Maumus M, Jorgensen C, Noël D. Therapeutic potential in rheumatic diseases of extracellular vesicles derived from mesenchymal stromal cells. Nat Rev Rheumatol 2023; 19:682-694. [PMID: 37666995 DOI: 10.1038/s41584-023-01010-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 09/06/2023]
Abstract
The incidence of rheumatic diseases such as rheumatoid arthritis and osteoarthritis and injuries to articular cartilage that lead to osteochondral defects is predicted to rise as a result of population ageing and the increase in high-intensity physical activities among young and middle-aged people. Current treatments focus on the management of pain and joint functionality to improve the patient's quality of life, but curative strategies are greatly desired. In the past two decades, the therapeutic value of mesenchymal stromal cells (MSCs) has been evaluated because of their regenerative potential, which is mainly attributed to the secretion of paracrine factors. Many of these factors are enclosed in extracellular vesicles (EVs) that reproduce the main functions of parental cells. MSC-derived EVs have anti-inflammatory, anti-apoptotic as well as pro-regenerative activities. Research on EVs has gained considerable attention as they are a potential cell-free therapy with lower immunogenicity and easier management than whole cells. MSC-derived EVs can rescue the pathogenetic phenotypes of chondrocytes and exert a protective effect in animal models of rheumatic disease. To facilitate the therapeutic use of EVs, appropriate cell sources for the production of EVs with the desired biological effects in each disease should be identified. Production and isolation of EVs should be optimized, and pre-isolation and post-isolation modifications should be considered to maximize the disease-modifying potential of the EVs.
Collapse
Affiliation(s)
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France.
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France.
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France.
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France.
| |
Collapse
|
31
|
Zhu T, Hu Z, Wang Z, Ding H, Li R, Wang J, Wang G. microRNA-301b-3p from mesenchymal stem cells-derived extracellular vesicles inhibits TXNIP to promote multidrug resistance of gastric cancer cells. Cell Biol Toxicol 2023; 39:1923-1937. [PMID: 35246762 DOI: 10.1007/s10565-021-09675-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) from mesenchymal stem cells (MSC)-derived extracellular vesicles (MSCs-EVs), including exosomes, are known to participate in different diseases. However, the function of miR-301b-3p from MSCs-EVs on the chemoresistance of gastric cancer (GC) cells remains poorly characterized. Thus, we aim to explore the role of MSCs-EVs-derived miR-301b-3p in multidrug resistance of GC cells. METHODS Cisplatin (DDP)/vincristine (VCR)-resistant and sensitive GC clinical samples were harvested to detect expression of miR-301b-3p and thioredoxin interacting protein (TXNIP). MSCs were respectively transfected with miR-301b-3p oligonucleotides and/or TXNIP plasmids to extract the EVs, which were then co-cultured with multidrug-resistant GC cells. Then, P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP), IC50, proliferation, migration, and apoptosis of resistant GC cells were determined. The tumor growth was observed in nude mice. Targeting relationship between miR-301b-3p and TXNIP was confirmed. RESULTS miR-301b-3p was upregulated, and TXNIP was downregulated in DDP/VCR-resistant GC tissues and cells. MSC-EVs induced drug resistance, proliferation, and migration and inhibited apoptosis of DDP/VCR-resistant GC cells in vitro, as well as facilitated tumor growth in vivo. Inhibition of miR-301b-3p or upregulation of TXNIP reversed the promoting effect of MSC-EVs on DDP/VCR resistant GC cells to DDP/VCR resistance and malignant behaviors. The effects of MSC-EVs carrying miR-301b-3p inhibition on DDP/VCR-resistant GC cells were reversed by TXNIP downregulation. TXNIP was confirmed as a target gene of miR-301b-3p. CONCLUSION miR-301b-3p from MSCs-EVs inhibits TXNIP to promote multidrug resistance of GC cells, providing a novel insight for chemotherapy in GC.
Collapse
Affiliation(s)
- Tianyu Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Zhihao Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Zhuoyin Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Hengxuan Ding
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Ruixin Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Jingtao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Guojun Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, No 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
32
|
Zhang Z, Zhao S, Sun Z, Zhai C, Xia J, Wen C, Zhang Y, Zhang Y. Enhancement of the therapeutic efficacy of mesenchymal stem cell-derived exosomes in osteoarthritis. Cell Mol Biol Lett 2023; 28:75. [PMID: 37770821 PMCID: PMC10540339 DOI: 10.1186/s11658-023-00485-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Osteoarthritis (OA), a common joint disorder with articular cartilage degradation as the main pathological change, is the major source of pain and disability worldwide. Despite current treatments, the overall treatment outcome is unsatisfactory. Thus, patients with severe OA often require joint replacement surgery. In recent years, mesenchymal stem cells (MSCs) have emerged as a promising therapeutic option for preclinical and clinical palliation of OA. MSC-derived exosomes (MSC-Exos) carrying bioactive molecules of the parental cells, including non-coding RNAs (ncRNAs) and proteins, have demonstrated a significant impact on the modulation of various physiological behaviors of cells in the joint cavity, making them promising candidates for cell-free therapy for OA. This review provides a comprehensive overview of the biosynthesis and composition of MSC-Exos and their mechanisms of action in OA. We also discussed the potential of MSC-Exos as a therapeutic tool for modulating intercellular communication in OA. Additionally, we explored bioengineering approaches to enhance MSC-Exos' therapeutic potential, which may help to overcome challenges and achieve clinically meaningful OA therapies.
Collapse
Affiliation(s)
- Zehao Zhang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Sheng Zhao
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhaofeng Sun
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Chuanxing Zhai
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| | - Yuge Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| |
Collapse
|
33
|
Wang Z, Zhu P, Liao B, You H, Cai Y. Effects and action mechanisms of individual cytokines contained in PRP on osteoarthritis. J Orthop Surg Res 2023; 18:713. [PMID: 37735688 PMCID: PMC10515001 DOI: 10.1186/s13018-023-04119-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Osteoarthritis (OA) is defined as a degenerative joint disease that can affect all tissues of the joint, including the articular cartilage, subchondral bone, ligaments capsule, and synovial membrane. The conventional nonoperative treatments are ineffective for cartilage repair and induce only symptomatic relief. Platelet-rich plasma (PRP) is a platelet concentrate derived from autologous whole blood with a high concentration of platelets, which can exert anti-inflammatory and regenerative effects by releasing multiple growth factors and cytokines. Recent studies have shown that PRP exhibits clinical benefits in patients with OA. However, high operational and equipment requirements greatly limit the application of PRP to OA treatment. Past studies have indicated that high-concentration PRP growth factors and cytokines may be applied as a commercial replacement for PRP. We reviewed the relevant articles to summarize the feasibility and mechanisms of PRP-based growth factors in OA. The available evidence suggests that transforming growth factor-α and β, platelet-derived growth factors, epidermal growth factor, insulin-like growth factor-1, and connective tissue growth factors might benefit OA, while vascular endothelial growth factor, tumor necrosis factor-α, angiopoietin-1, and stromal cell derived factor-1α might induce negative effects on OA. The effects of fibroblast growth factor, hepatocyte growth factor, platelet factor 4, and keratinocyte growth factor on OA remain uncertain. Thus, it can be concluded that not all cytokines released by PRP are beneficial, although the therapeutic action of PRP has a valuable potential to improve.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan, China
| | - Pengfei Zhu
- Department of Cardiovascular, Wuhan Fourth Hospital, Wuhan, China
- Department of Cardiovascular, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bokai Liao
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University and Technology, Jiefang Avenue No.1095, Qiaokou District, Wuhan, 430030, Hubei Province, China.
| | - Yu Cai
- Department of Rehabilitation, Wuhan Fourth Hospital, Hanzheng Street No.473, Qiaokou District, Wuhan, 430000, Hubei Province, China.
| |
Collapse
|
34
|
Chen R, Pye JS, Li J, Little CB, Li JJ. Multiphasic scaffolds for the repair of osteochondral defects: Outcomes of preclinical studies. Bioact Mater 2023; 27:505-545. [PMID: 37180643 PMCID: PMC10173014 DOI: 10.1016/j.bioactmat.2023.04.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/18/2023] [Accepted: 04/17/2023] [Indexed: 05/16/2023] Open
Abstract
Osteochondral defects are caused by injury to both the articular cartilage and subchondral bone within skeletal joints. They can lead to irreversible joint damage and increase the risk of progression to osteoarthritis. Current treatments for osteochondral injuries are not curative and only target symptoms, highlighting the need for a tissue engineering solution. Scaffold-based approaches can be used to assist osteochondral tissue regeneration, where biomaterials tailored to the properties of cartilage and bone are used to restore the defect and minimise the risk of further joint degeneration. This review captures original research studies published since 2015, on multiphasic scaffolds used to treat osteochondral defects in animal models. These studies used an extensive range of biomaterials for scaffold fabrication, consisting mainly of natural and synthetic polymers. Different methods were used to create multiphasic scaffold designs, including by integrating or fabricating multiple layers, creating gradients, or through the addition of factors such as minerals, growth factors, and cells. The studies used a variety of animals to model osteochondral defects, where rabbits were the most commonly chosen and the vast majority of studies reported small rather than large animal models. The few available clinical studies reporting cell-free scaffolds have shown promising early-stage results in osteochondral repair, but long-term follow-up is necessary to demonstrate consistency in defect restoration. Overall, preclinical studies of multiphasic scaffolds show favourable results in simultaneously regenerating cartilage and bone in animal models of osteochondral defects, suggesting that biomaterials-based tissue engineering strategies may be a promising solution.
Collapse
Affiliation(s)
- Rouyan Chen
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney, NSW, 2065, Australia
- School of Electrical and Mechanical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, SA, 5005, Australia
| | - Jasmine Sarah Pye
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia
| | - Jiarong Li
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney, NSW, 2065, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia
| | - Christopher B. Little
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney, NSW, 2065, Australia
| | - Jiao Jiao Li
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney, NSW, 2065, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia
| |
Collapse
|
35
|
Ossendorff R, Grad S, Tertel T, Wirtz DC, Giebel B, Börger V, Schildberg FA. Immunomodulatory potential of mesenchymal stromal cell-derived extracellular vesicles in chondrocyte inflammation. Front Immunol 2023; 14:1198198. [PMID: 37564645 PMCID: PMC10410457 DOI: 10.3389/fimmu.2023.1198198] [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: 03/31/2023] [Accepted: 07/03/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction Osteoarthritis (OA) affects a large percentage of the population worldwide. Current surgical and nonsurgical concepts for treating OA only result in symptom-modifying effects. However, there is no disease-modifying therapy available. Extracellular vesicles released by mesenchymal stem/stromal cells (MSC-EV) are promising agents to positively influence joint homeostasis in the osteoarthritic surroundings. This pilot study aimed to investigate the effect of characterized MSC-EVs on chondrogenesis in a 3D chondrocyte inflammation model with the pro-inflammatory cytokine TNFα. Methods Bovine articular chondrocytes were expanded and transferred into pellet culture at passage 3. TNFα, human MSC-EV preparations (MSC-EV batches 41.5-EVi1 and 84-EVi), EVs from human platelet lysate (hPL4-EV), or the combination of TNFα and EVs were supplemented. To assess the effect of MSC-EVs in the chondrocyte inflammation model after 14 days, DNA, glycosaminoglycan (GAG), total collagen, IL-6, and NO release were quantified, and gene expression of anabolic (COL-II, aggrecan, COMP, and PRG-4), catabolic (MMP-3, MMP-13, ADAMTS-4 and ADAMTS-5), dedifferentiation (COL-I), hypertrophy (COL-X, VEGF), and inflammatory (IL-8) markers were analyzed; histological evaluation was performed using safranin O/Fast Green staining and immunohistochemistry of COL I and II. For statistical evaluation, nonparametric tests were chosen with a significance level of p < 0.05. Results TNFα supplementation resulted in catabolic stimulation with increased levels of NO and IL-6, upregulation of catabolic gene expression, and downregulation of anabolic markers. These findings were supported by a decrease in matrix differentiation (COL-II). Supplementation of EVs resulted in an upregulation of the chondrogenic marker PRG-4. All MSC-EV preparations significantly increased GAG retention per pellet. In contrast, catabolic markers and IL-8 expression were upregulated by 41.5-EVi1. Regarding protein levels, IL-6 and NO release were increased by 41.5-EVi1. Histologic and immunohistochemical evaluations indicated a higher differentiation potential of chondrocytes treated with 84-EVi. Discussion MSC-EVs can positively influence chondrocyte matrix production in pro-inflammatory surroundings, but can also stimulate inflammation. In this study MSC-EV 41.5-EVi1 supplementation increased chondrocyte inflammation, whereas MSC-84-EVi supplementation resulted a higher chondrogenic potential of chondrocytes in 3D pellet culture. In summary, the selected MSC-EVs exhibited promising chondrogenic effects indicating their significant potential for the treatment of OA; however, the functional heterogeneity in MSC-EV preparations has to be solved.
Collapse
Affiliation(s)
- Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Verena Börger
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
36
|
Mishra A, Kumar R, Mishra SN, Vijayaraghavalu S, Tiwari NK, Shukla GC, Gurusamy N, Kumar M. Differential Expression of Non-Coding RNAs in Stem Cell Development and Therapeutics of Bone Disorders. Cells 2023; 12:cells12081159. [PMID: 37190068 PMCID: PMC10137108 DOI: 10.3390/cells12081159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Stem cells' self-renewal and multi-lineage differentiation are regulated by a complex network consisting of signaling factors, chromatin regulators, transcription factors, and non-coding RNAs (ncRNAs). Diverse role of ncRNAs in stem cell development and maintenance of bone homeostasis have been discovered recently. The ncRNAs, such as long non-coding RNAs, micro RNAs, circular RNAs, small interfering RNA, Piwi-interacting RNAs, etc., are not translated into proteins but act as essential epigenetic regulators in stem cells' self-renewal and differentiation. Different signaling pathways are monitored efficiently by the differential expression of ncRNAs, which function as regulatory elements in determining the fate of stem cells. In addition, several species of ncRNAs could serve as potential molecular biomarkers in early diagnosis of bone diseases, including osteoporosis, osteoarthritis, and bone cancers, ultimately leading to the development of new therapeutic strategies. This review aims to explore the specific roles of ncRNAs and their effective molecular mechanisms in the growth and development of stem cells, and in the regulation of osteoblast and osteoclast activities. Furthermore, we focus on and explore the association of altered ncRNA expression with stem cells and bone turnover.
Collapse
Affiliation(s)
- Anurag Mishra
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Rishabh Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Satya Narayan Mishra
- Maa Gayatri College of Pharmacy, Dr. APJ Abdul Kalam Technical University, Prayagraj 211009, India
| | | | - Neeraj Kumar Tiwari
- Department of IT-Satellite Centre, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India
| | - Girish C Shukla
- Department of Biological, Geological, and Environmental Sciences, 2121 Euclid Ave., Cleveland, OH 44115, USA
- Center for Gene Regulation in Health and Disease, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Munish Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| |
Collapse
|
37
|
Schrenker S, Cucchiarini M, Goebel L, Oláh T, Venkatesan JK, Schmitt G, Speicher-Mentges S, Maihöfer J, Gao L, Zurakowski D, Menger MD, Laschke MW, Madry H. In vivo rAAV-mediated human TGF-β overexpression reduces perifocal osteoarthritis and improves osteochondral repair in a large animal model at one year. Osteoarthritis Cartilage 2023; 31:467-481. [PMID: 36481450 DOI: 10.1016/j.joca.2022.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/20/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Osteoarthritis (OA) is a serious consequence of focal osteochondral defects. Gene transfer of human transforming growth factor beta (hTGF-β) with recombinant adeno-associated virus (rAAV) vectors offers a strategy to improve osteochondral repair. However, the long-term in vivo effects of such rAAV-mediated TGF-β overexpression including its potential benefits on OA development remain unknown. METHOD Focal osteochondral defects in minipig knees received rAAV-lacZ (control) or rAAV-hTGF-β in vivo. After one year, osteochondral repair and perifocal OA were visualized using validated macroscopic scoring, ultra-high-field MRI at 9.4 T, and micro-CT. A quantitative estimation of the cellular densities and a validated semi-quantitative scoring of histological and immunohistological parameters completed the analysis of microarchitectural parameters. RESULTS Direct rAAV-hTGF-β application induced and maintained significantly improved defect filling and safranin O staining intensity and overall cartilage repair at one year in vivo. In addition, rAAV-hTGF-β led to significantly higher chondrocyte densities within the cartilaginous repair tissue without affecting chondrocyte hypertrophy and minimized subarticular trabecular separation. Of note, rAAV-hTGF-β significantly improved the adjacent cartilage structure and chondrocyte density and reduced overall perifocal OA development after one year in vivo. CONCLUSIONS rAAV-hTGF-β treatment improves long-term osteochondral repair and delays the progression of perifocal OA in a translational model. These findings have considerable potential for targeted molecular approaches to treat focal osteochondral defects.
Collapse
Affiliation(s)
- S Schrenker
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - M Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - L Goebel
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - T Oláh
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - J K Venkatesan
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - G Schmitt
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - S Speicher-Mentges
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - J Maihöfer
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - L Gao
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - D Zurakowski
- Departments of Anesthesia and Surgery, Children's Hospital Boston, Harvard Medical School, Boston, MA, 02115, USA.
| | - M D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| | - M W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| | - H Madry
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| |
Collapse
|
38
|
van de Looij SM, de Jong OG, Vermonden T, Lorenowicz MJ. Injectable hydrogels for sustained delivery of extracellular vesicles in cartilage regeneration. J Control Release 2023; 355:685-708. [PMID: 36739906 DOI: 10.1016/j.jconrel.2023.01.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/15/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are a population of small vesicles secreted by essentially all cell types, containing a wide variety of biological macromolecules. Due to their intrinsic capabilities for efficient intercellular communication, they are involved in various aspects of cellular functioning. In the past decade, EVs derived from stem cells attracted interest in the field of regenerative medicine. Owing to their regenerative properties, they have great potential for use in tissue repair, in particular for tissues with limited regenerative capabilities such as cartilage. The maintenance of articular cartilage is dependent on a precarious balance of many different components that can be disrupted by the onset of prevalent rheumatic diseases. However, while cartilage is a tissue with strong mechanical properties that can withstand movement and heavy loads for years, it is virtually incapable of repairing itself after damage has occurred. Stem cell-derived EVs (SC-EVs) transport regenerative components such as proteins and nucleic acids from their parental cells to recipient cells, thereby promoting cartilage healing. Many possible pathways through which SC-EVs execute their regenerative function have been reported, but likely there are still numerous other pathways that are still unknown. This review discusses various preclinical studies investigating intra-articular injections of free SC-EVs, which, while often promoting chondrogenesis and cartilage repair in vivo, showed a recurring limitation of the need for multiple administrations to achieve sufficient tissue regeneration. Potentially, this drawback can be overcome by making use of an EV delivery platform that is capable of sustainably releasing EVs over time. With their remarkable versatility and favourable chemical, biological and mechanical properties, hydrogels can facilitate this release profile by encapsulating EVs in their porous structure. Ideally, the optimal delivery platform can be formed in-situ, by means of an injectable hydrogel that can be administered directly into the affected joint. Relevant research fulfilling these criteria is discussed in detail, including the steps that still need to be taken before injectable hydrogels for sustained delivery of EVs can be applied in the context of cartilage regeneration in the clinic.
Collapse
Affiliation(s)
- Sanne M van de Looij
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Olivier G de Jong
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Magdalena J Lorenowicz
- Regenerative Medicine Centre, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands; Centre for Molecular Medicine, University Medical Centre Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands; Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands.
| |
Collapse
|
39
|
You B, Zhou C, Yang Y. MSC-EVs alleviate osteoarthritis by regulating microenvironmental cells in the articular cavity and maintaining cartilage matrix homeostasis. Ageing Res Rev 2023; 85:101864. [PMID: 36707035 DOI: 10.1016/j.arr.2023.101864] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
Osteoarthritis (OA), a common cause of chronic articular cartilage degeneration, is the main cause of disability in older adults and severely affects quality of life. Multiple factors are involved in the pathogenesis of OA, resulting in imbalance in the homeostasis of the joint cavity microenvironment, which exacerbates the disease. Because of the deficiency of blood vessels and nerves in cartilage, existing therapies to promote cartilage healing are relatively ineffective. Mesenchymal stem cell (MSC)-related therapies have achieved positive outcomes for the treatment of OA, and these beneficial effects have been confirmed to be largely mediated by extracellular vesicles (EVs). MSC-derived EVs (MSC-EVs) have been demonstrated to participate in the regulation of chondrocyte function, to have anti-inflammatory and immunomodulatory effects, and to alleviate metabolic disorders of the extracellular matrix, thereby slowing the progression of OA. In addition, engineered MSC-EVs can enrich therapeutic molecules and optimize administration to enhance their therapeutic effects on OA. A thorough understanding of the endogenous properties of EVs and related engineering strategies could help researchers develop more precise control therapy for OA.
Collapse
Affiliation(s)
- Benshuai You
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Chenglin Zhou
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| | - Yang Yang
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| |
Collapse
|
40
|
Gerami MH, Khorram R, Rasoolzadegan S, Mardpour S, Nakhaei P, Hashemi S, Al-Naqeeb BZT, Aminian A, Samimi S. Emerging role of mesenchymal stem/stromal cells (MSCs) and MSCs-derived exosomes in bone- and joint-associated musculoskeletal disorders: a new frontier. Eur J Med Res 2023; 28:86. [PMID: 36803566 PMCID: PMC9939872 DOI: 10.1186/s40001-023-01034-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 01/26/2023] [Indexed: 02/22/2023] Open
Abstract
Exosomes are membranous vesicles with a 30 to 150 nm diameter secreted by mesenchymal stem/stromal cells (MSCs) and other cells, such as immune cells and cancer cells. Exosomes convey proteins, bioactive lipids, and genetic components to recipient cells, such as microRNAs (miRNAs). Consequently, they have been implicated in regulating intercellular communication mediators under physiological and pathological circumstances. Exosomes therapy as a cell-free approach bypasses many concerns regarding the therapeutic application of stem/stromal cells, including undesirable proliferation, heterogeneity, and immunogenic effects. Indeed, exosomes have become a promising strategy to treat human diseases, particularly bone- and joint-associated musculoskeletal disorders, because of their characteristics, such as potentiated stability in circulation, biocompatibility, low immunogenicity, and toxicity. In this light, a diversity of studies have indicated that inhibiting inflammation, inducing angiogenesis, provoking osteoblast and chondrocyte proliferation and migration, and negative regulation of matrix-degrading enzymes result in bone and cartilage recovery upon administration of MSCs-derived exosomes. Notwithstanding, insufficient quantity of isolated exosomes, lack of reliable potency test, and exosomes heterogeneity hurdle their application in clinics. Herein, we will deliver an outline respecting the advantages of MSCs-derived exosomes-based therapy in common bone- and joint-associated musculoskeletal disorders. Moreover, we will have a glimpse the underlying mechanism behind the MSCs-elicited therapeutic merits in these conditions.
Collapse
Affiliation(s)
- Mohammad Hadi Gerami
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roya Khorram
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheil Rasoolzadegan
- grid.411600.2Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Mardpour
- grid.411705.60000 0001 0166 0922Department of Radiology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooria Nakhaei
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheyla Hashemi
- grid.411036.10000 0001 1498 685XObstetrician, Gynaecology & Infertility Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Amir Aminian
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Sahar Samimi
- Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Li J, Zhao B, Yao S, Dai Y, Zhang X, Yang N, Bao Z, Cai J, Chen Y, Wu X. Dermal PapillaCell-Derived Exosomes Regulate Hair Follicle Stem Cell Proliferation via LEF1. Int J Mol Sci 2023; 24:3961. [PMID: 36835374 PMCID: PMC9964005 DOI: 10.3390/ijms24043961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/06/2023] [Accepted: 02/11/2023] [Indexed: 02/18/2023] Open
Abstract
Hair follicle (HF) growth and development are controlled by various cell types, including hair follicle stem cells (HFSCs) and dermal papilla cells (DPCs). Exosomes are nanostructures that participate in many biological processes. Accumulating evidence indicates that DPC-derived exosomes (DPC-Exos) mediate HFSC proliferation and differentiation during the cyclical growth of hair follicles. In this study, we found that DPC-Exos increase ki67 expression and CCK8 cell viability readouts in HFSCs but reduce annexin staining of apoptotic cells. RNA sequencing of DPC-Exos-treated HFSCs identified 3702 significantly differentially expressed genes (DEGs), including BMP4, LEF1, IGF1R, TGFβ3, TGFα, and KRT17. These DEGs were enriched in HF growth- and development-related pathways. We further verified the function of LEF1 and showed that overexpression of LEF1 increased the expression of HF development-related genes and proteins, enhanced HFSC proliferation, and reduced HFSC apoptosis, while knockdown of LEF1 reversed these effects. DPC-Exos could also rescue the siRNA-LEF1 effect in HFSCs. In conclusion, this study demonstrates that DPC-Exos mediated cell-to-cell communication can regulate HFSCs proliferation by stimulating LEF1 and provide novel insights into HF growth and development regulatory mechanisms.
Collapse
Affiliation(s)
- Jiali Li
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Bohao Zhao
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Shuyu Yao
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Yingying Dai
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Xiyu Zhang
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Naisu Yang
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Zhiyuan Bao
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Jiawei Cai
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Yang Chen
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| | - Xinsheng Wu
- College of Animal Science and Technology, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, 48 South University Ave Yangzhou, Yangzhou 225009, China
| |
Collapse
|
42
|
Li J, Wang ZH, Sun YH. TGF-β1 stimulated mesenchymal stem cells-generated exosomal miR-29a promotes the proliferation, migration and fibrogenesis of tenocytes by targeting FABP3. Cytokine 2023; 162:156090. [PMID: 36481477 DOI: 10.1016/j.cyto.2022.156090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 10/30/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Rotator cuff Tear (RCT) causes a lot of inconvenience for patients. In most cases, RCT injury does not heal back to bone after repair, and there is a high chance of retearing. Therefore, there is a need to explore more effective targeted therapies. Bone mesenchymal stem cell-derived exosome (BMSCs-Exo) has been proved to be beneficial to the proliferation of tendon cells, but its specific mechanism remains to be further explored. METHODS BMSCs-Exo was isolated and identified by detecting the specific markers using flow cytometry and western blot assays. qRT-PCR and western blot were utilized to determine the gene or protein expressions, respectively. Cell proliferation, and migration in tenocytes were measured by CCK8, EdU and transwell assays. The interaction between miR-29a and FABP3 was analyzed using dual-luciferase reporter assay. RESULTS Our findings demonstrated that miR-29a was expressed in BMSCs-Exo and could be significantly enriched after TGF-β1 treatment. Moreover, TGF-β1-modified BMSCs-Exo co-cultured could promote the proliferation, migration and fibrosis of tenocytes by carrying miR-29a. Upon miR-29a was reduced in BMSCs-Exo, the regulatory roles of BMSCs-Exo on tenocytes were reversed. Mechanistically, miR-29a negatively regulated FABP3 via interaction with its 3'-UTR. Enforced expression of FABP3 could reverse the modulation of exosomal miR-29a in tenocytes. CONCLUSION Exosomal miR-29a derived from TGF-β1-modified BMSCs facilitated the proliferation, migration and fibrosis of tenocytes through targeting FABP3.
Collapse
Affiliation(s)
- Jia Li
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, PR China.
| | - Zhi-Hui Wang
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, PR China
| | - Yu-Hang Sun
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, PR China
| |
Collapse
|
43
|
Exosomes treating osteoarthritis: hope with challenge. Heliyon 2023; 9:e13152. [PMID: 36711315 PMCID: PMC9880404 DOI: 10.1016/j.heliyon.2023.e13152] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/23/2023] Open
Abstract
Osteoarthritis (OA) has been proven as the second primary cause of pain and disability in the elderly population, impact patients both physically and mentally, as well as imposing a heavy burden on the global healthcare system. Current treatment methods, whether conservative or surgical, that aim at relieving symptoms can not delay or reverse the degenerative process in the structure. Scientists and clinicians are facing a revolution in OA treatment strategies. The emergence of exosomes brings hope for OA treatment based on pathology, which is attributed to its full potential in protecting chondrocytes from excessive death, alleviating inflammation, maintaining cartilage matrix metabolism, and regulating angiogenesis and subchondral bone remodeling. Therefore, we summarized the recent studies of exosomes in OA, aiming to comprehensively understand the functions and mechanisms of exosomes in OA treatment, which may provide direction and theoretical support for formulating therapeutic strategies in the future.
Collapse
|
44
|
Li F, Xu Z, Xie Z, Sun X, Li C, Chen Y, Xu J, Pi G. Adipose mesenchymal stem cells-derived exosomes alleviate osteoarthritis by transporting microRNA -376c-3p and targeting the WNT-beta-catenin signaling axis. Apoptosis 2022; 28:362-378. [PMID: 36396777 DOI: 10.1007/s10495-022-01787-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
Abstract
Osteoarthritis (OA), one of the major diseases afflicting the elderly, is a type of degenerative joint disease related to cartilage and synovium. This study aimed to clarify the role and mechanism of adipose mesenchymal stem cell (ADSC)-derived exosomes (Exos) in OA-induced chondrocyte degradation and synovial hyperplasia, thus improving the quality of life of patients. The rat OA model, chondrocytes, synovial fibroblast models and immunofluorescence were applied to observe the in vivo and in vitro functions of human ADSC (hADSC)-derived Exos in OA and its possible regulatory signaling pathways. Bioinformatics software and luciferase reporter assay were carried out to verify the mechanism of microRNA-376c-3p (miR-376c-3p) in hADSC-derived Exos in OA in vitro. Moreover, Safranine O-Fast Green Cartilage staining, Masson staining, immunohistochemistry and immunofluorescence were conducted to verify the role of miR-376c-3p in hADSC-derived Exos in OA in vivo. hADSC-derived Exos mitigated OA-induced chondrocyte degradation and synovial fibrosis both in vivo and in vitro models by repressing the WNT-beta-catenin signaling pathway. For the mechanism exploration in vitro, miR-376c-3p was raised in hADSC-derived Exos and mediated the fibrosis of synovial fibroblasts in OA, and miR-376c-3p targeted the 3'-untranslated region of WNT3 or WNT9a. Meanwhile, the in vivo experiments also corroborated that the miR-376c-3p in hADSC-derived Exos mitigated OA-induced chondrocyte degradation and synovial fibrosis. MiR-376c-3p in hADSC-derived Exos repressed the WNT-beta-catenin pathway by targeting WNT3 or WNT9a, and then mitigating OA-induced chondrocyte degradation and synovial fibrosis, thereby providing theoretical basis for clinical implementation of treatment.
Collapse
Affiliation(s)
- Feng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Zhiming Xu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Zheng Xie
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xing Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Chengxiang Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yangyang Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Jianzhong Xu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Guofu Pi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
45
|
MSC-EV therapy for bone/cartilage diseases. Bone Rep 2022; 17:101636. [DOI: 10.1016/j.bonr.2022.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
|
46
|
Li XC, Luo SJ, Fan W, Zhou TL, Huang CM, Wang MS. M2 macrophage-conditioned medium inhibits intervertebral disc degeneration in a tumor necrosis factor-α-rich environment. J Orthop Res 2022; 40:2488-2501. [PMID: 35170802 DOI: 10.1002/jor.25292] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/26/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023]
Abstract
Inflammation is the primary pathological phenomenon associated with disc degeneration; the inflammatory cytokine tumor necrosis factor (TNF-α) plays a crucial role in this pathology. The anti-inflammatory and regenerative effects of M2 macrophages on nucleus pulposus cells (NPCs) in intervertebral disc degeneration (IDD) progression remain unknown. Here, M2 conditioned medium (M2CM) was harvested and purified from human acute monocytic leukaemia cell line (THP-1) cells and mouse peritoneal macrophages, respectively; it was used for culturing human NPCs and a mouse intervertebral disc (IVD) organ culture model. NPCs and IVD organ models were divided into three groups: group 1 treated with 10% fetal bovine serum (control); group 2 treated with 10 ng/ml TNF-α; and group 3 treated with 10 ng/ml TNF-α and M2CM (coculture group). After 2-14 days, cell proliferation, extracellular matrix synthesis, apoptosis, and NPC senescence were assessed. Cell proliferation was reduced in TNF-α-treated NPCs and inhibited in the M2CM co-culture treatment. Moreover, TNF-α treatment enhanced apoptosis, senescence, and expression of inflammatory factor-related genes, including interleukin-6, MMP-13, ADAMTS-4, and ADAMTS-5, whereas M2CM coculture significantly reversed these effects. In addition, co-culture with M2CM promoted aggrecan and collagen II synthesis, but reduced collagen Iα1 levels in TNF-α treatment groups. Using our established three-dimensional murine IVD organ culture model, we show that M2CM suppressed the inhibitory effect of TNF-α-rich environment. Therefore, co-culture with M2CM promotes cell proliferation and extracellular matrix synthesis and inhibits inflammation, apoptosis, and NPC senescence. This study highlights the therapeutic potential of M2CM for IDD.
Collapse
Affiliation(s)
- Xiao-Chuan Li
- Postdoctoral Innovation Practice Base of Gaozhou People's Hospital, Gaozhou People's Hospital, Maoming, Guangdong, China.,Department of Cell Biology, Southern Medical University, Guangzhou, China.,Department of Orthopedic Surgery, Gaozhou People's Hospital, Maoming, Guangdong, China
| | - Shao-Jian Luo
- Department of Orthopedic Surgery, Gaozhou People's Hospital, Maoming, Guangdong, China
| | - Wu Fan
- Department of Orthopedic Surgery, Gaozhou People's Hospital, Maoming, Guangdong, China
| | - Tian-Li Zhou
- Department of Orthopedic Surgery, Gaozhou People's Hospital, Maoming, Guangdong, China
| | - Chun-Ming Huang
- Postdoctoral Innovation Practice Base of Gaozhou People's Hospital, Gaozhou People's Hospital, Maoming, Guangdong, China.,Department of Orthopedic Surgery, Gaozhou People's Hospital, Maoming, Guangdong, China
| | - Mao-Sheng Wang
- Postdoctoral Innovation Practice Base of Gaozhou People's Hospital, Gaozhou People's Hospital, Maoming, Guangdong, China.,Department of Cell Biology, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Yin H, Li M, Tian G, Ma Y, Ning C, Yan Z, Wu J, Ge Q, Sui X, Liu S, Zheng J, Guo W, Guo Q. The role of extracellular vesicles in osteoarthritis treatment via microenvironment regulation. Biomater Res 2022; 26:52. [PMID: 36199125 PMCID: PMC9532820 DOI: 10.1186/s40824-022-00300-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/18/2022] [Indexed: 11/10/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that is common among the middle-aged and older populations, causes patients to experience recurrent pain in their joints and negatively affects their quality of life. Currently, therapeutic options for patients with OA consist of medications to alleviate pain and treat the symptoms; however, due to typically poor outcomes, patients with advanced OA are unlikely to avoid joint replacement. In recent years, several studies have linked disrupted homeostasis of the joint cavity microenvironment to the development of OA. Recently, extracellular vesicles (EVs) have received increasing attention in the field of OA. EVs are natural nano-microcarrier materials with unique biological activity that are produced by cells through paracrine action. They are composed of lipid bilayers that contain physiologically active molecules, such as nucleic acids and proteins. Moreover, EVs may participate in local and distal intercellular and intracellular communication. EVs have also recently been shown to influence OA development by regulating biochemical factors in the OA microenvironmental. In this article, we first describe the microenvironment of OA. Then, we provide an overview of EVs, summarize the main types used for the treatment of OA, and describe their mechanisms. Next, we review clinical studies using EVs for OA treatment. Finally, the specific mechanism underlying the application of miRNA-enriched EVs in OA therapy is described.
Collapse
Affiliation(s)
- Han Yin
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Muzhe Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
- Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, 421000, China
| | - Guangzhao Tian
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yang Ma
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Chao Ning
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Zineng Yan
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Jiang Wu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Qian Ge
- Huaiyin People's Hospital of Huai'an, Huai'an, 223001, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China.
| | - Jinxuan Zheng
- Department of Orthodontics, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, No.56 Linyuan Xi Road, Yuexiu District, Guangzhou, Guangdong, 510055, People's Republic of China.
| | - Weimin Guo
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Second Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China.
| |
Collapse
|
48
|
Jeyaraman M, Muthu S, Shehabaz S, Jeyaraman N, Rajendran RL, Hong CM, Nallakumarasamy A, Packkyarathinam RP, Sharma S, Ranjan R, Khanna M, Ahn BC, Gangadaran P. Current understanding of MSC-derived exosomes in the management of knee osteoarthritis. Exp Cell Res 2022; 418:113274. [PMID: 35810774 DOI: 10.1016/j.yexcr.2022.113274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) have been utilized as medicinal agents or as delivery vehicles in cartilage injuries and cartilage-based diseases. Given the ongoing emergence of evidence on the effector mechanisms and methods of the utility of the MSC-Exos in knee osteoarthritis, a comprehensive review of the current evidence is the need of the hour. Hence, in this article, we review the current understanding of the role of MSC-Exos in the management of knee osteoarthritis in view of their classification, characterization, biogenesis, mechanism of action, pathways involved in their therapeutic action, in-vitro evidence on cartilage regeneration, in-vivo evidence in OA knee models and recent advances in using MSC-Exos to better streamline future research from bench to bedside for OA knee.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine - Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, 600095, Tamil Nadu, India; Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, 201310, Uttar Pradesh, India; Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, 201310, Uttar Pradesh, India; Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Department of Orthopaedics, Government Medical College and Hospital, Dindigul, 624304, Tamil Nadu, India
| | - Syed Shehabaz
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow, 226010, Uttar Pradesh, India
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow, 226010, Uttar Pradesh, India; Joint Replacement, Department of Orthopaedics, Atlas Hospitals, Tiruchirappalli, 620002, Tamil Nadu, India.
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar, 751019, Odissa, India
| | | | - Shilpa Sharma
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, 201310, Uttar Pradesh, India
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Department of Orthopaedics, Prasad Institute of Medical Sciences, Lucknow, 226401, Uttar Pradesh, India
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
49
|
Kim HY, Kwon S, Um W, Shin S, Kim CH, Park JH, Kim BS. Functional Extracellular Vesicles for Regenerative Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106569. [PMID: 35322545 DOI: 10.1002/smll.202106569] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/22/2022] [Indexed: 06/14/2023]
Abstract
The unique biological characteristics and promising clinical potential of extracellular vesicles (EVs) have galvanized EV applications for regenerative medicine. Recognized as important mediators of intercellular communication, naturally secreted EVs have the potential, as innate biotherapeutics, to promote tissue regeneration. Although EVs have emerged as novel therapeutic agents, challenges related to the clinical transition have led to further functionalization. In recent years, various engineering approaches such as preconditioning, drug loading, and surface modification have been developed to potentiate the therapeutic outcomes of EVs. Also, limitations of natural EVs have been addressed by the development of artificial EVs that offer advantages in terms of production yield and isolation methodologies. In this review, an updated overview of current techniques is provided for the functionalization of natural EVs and recent advances in artificial EVs, particularly in the scope of regenerative medicine.
Collapse
Affiliation(s)
- Han Young Kim
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Seunglee Kwon
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Wooram Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sol Shin
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Interdisciplinary Program of Bioengineering, Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
50
|
Zhang L, Lin Y, Zhang X, Shan C. Research progress of exosomes in orthopedics. Front Genet 2022; 13:915141. [PMID: 36081990 PMCID: PMC9445804 DOI: 10.3389/fgene.2022.915141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022] Open
Abstract
Exosomes are nano-extracellular vesicles secreted by a variety of cells. They are composed of a double-layer membrane that can transport a variety of proteins, coding and non-coding genes, and bioactive substances. Exosomes participate in information transmission between cells and regulate processes such as cell proliferation, migration, angiogenesis, and phenotypic transformation. They have broad prospects in the occurrence, development, and treatment of many diseases including orthopedics. Exosomes derived from different types of bone cells such as mesenchymal stem cells, osteoblasts, osteoclasts, and their precursors are recognized to play pivotal roles in bone remodeling processes including osteogenesis, osteoclastogenesis, and angiogenesis. This articlesummarizes the characteristics of exosomes and their research progress in bone remodeling, bone tumors, vascular skeletal muscle injury, spinal cord injury, degenerative disc diseases, cartilage degeneration, osteoarthritis, necrosis of the femoral head, and osteoporosis.
Collapse
|