101
|
Davies OG. Extracellular vesicles: From bone development to regenerative orthopedics. Mol Ther 2023; 31:1251-1274. [PMID: 36869588 PMCID: PMC10188641 DOI: 10.1016/j.ymthe.2023.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Regenerative medicine aims to promote the replacement of tissues lost to damage or disease. While positive outcomes have been observed experimentally, challenges remain in their clinical translation. This has led to growing interest in applying extracellular vesicles (EVs) to augment or even replace existing approaches. Through the engineering of culture environments or direct/indirect manipulation of EVs themselves, multiple avenues have emerged to modulate EV production, targeting, and therapeutic potency. Drives to modulate release using material systems or functionalize implants for improved osseointegration have also led to outcomes that could have real-world impact. The purpose of this review is to highlight advantages in applying EVs for the treatment of skeletal defects, outlining the current state of the art in the field and emphasizing avenues for further investigation. Notably, the review identifies inconsistencies in EV nomenclature and outstanding challenges in defining a reproducible therapeutic dose. Challenges also remain in the scalable manufacture of a therapeutically potent and pure EV product, with a need to address scalable cell sources and optimal culture environments. Addressing these issues will be critical if we are to develop regenerative EV therapies that meet the demands of regulators and can be translated from bench to bedside.
Collapse
Affiliation(s)
- Owen G Davies
- School of Sport, Exercise, and Health Sciences, Loughborough University, Epinal Way, Loughborough, Leicestershire LE11 3TU, UK.
| |
Collapse
|
102
|
Mélou C, Pellen-Mussi P, Novello S, Brézulier D, Novella A, Tricot S, Bellaud P, Chauvel-Lebret D. Spheroid Culture System, a Promising Method for Chondrogenic Differentiation of Dental Mesenchymal Stem Cells. Biomedicines 2023; 11:biomedicines11051314. [PMID: 37238984 DOI: 10.3390/biomedicines11051314] [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: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
The objective of the present work was to develop a three-dimensional culture model to evaluate, in a short period of time, cartilage tissue engineering protocols. The spheroids were compared with the gold standard pellet culture. The dental mesenchymal stem cell lines were from pulp and periodontal ligament. The evaluation used RT-qPCR and Alcian Blue staining of the cartilage matrix. This study showed that the spheroid model allowed for obtaining greater fluctuations of the chondrogenesis markers than for the pellet one. The two cell lines, although originating from the same organ, led to different biological responses. Finally, biological changes were detectable for short periods of time. In summary, this work demonstrated that the spheroid model is a valuable tool for studying chondrogenesis and the mechanisms of osteoarthritis, and evaluating cartilage tissue engineering protocols.
Collapse
Affiliation(s)
- Caroline Mélou
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
- UFR Odontologie, University of Rennes, 35043 Rennes, France
| | - Pascal Pellen-Mussi
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
| | - Solen Novello
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
- UFR Odontologie, University of Rennes, 35043 Rennes, France
| | - Damien Brézulier
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
| | - Agnès Novella
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
| | - Sylvie Tricot
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
| | - Pascale Bellaud
- CNRS, Inserm UMS Biosit, France BioImaging, Core Facility H2P2, University of Rennes, 35000 Rennes, France
| | - Dominique Chauvel-Lebret
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
- UFR Odontologie, University of Rennes, 35043 Rennes, France
| |
Collapse
|
103
|
Ong HS, Riau AK, Yam GHF, Yusoff NZBM, Han EJY, Goh TW, Lai RC, Lim SK, Mehta JS. Mesenchymal Stem Cell Exosomes as Immunomodulatory Therapy for Corneal Scarring. Int J Mol Sci 2023; 24:7456. [PMID: 37108619 PMCID: PMC10144287 DOI: 10.3390/ijms24087456] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Corneal scarring is a leading cause of worldwide blindness. Human mesenchymal stem cells (MSC) have been reported to promote corneal wound healing through secreted exosomes. This study investigated the wound healing and immunomodulatory effects of MSC-derived exosomes (MSC-exo) in corneal injury through an established rat model of corneal scarring. After induction of corneal scarring by irregular phototherapeutic keratectomy (irrPTK), MSC exosome preparations (MSC-exo) or PBS vehicle as controls were applied to the injured rat corneas for five days. The animals were assessed for corneal clarity using a validated slit-lamp haze grading score. Stromal haze intensity was quantified using in-vivo confocal microscopy imaging. Corneal vascularization, fibrosis, variations in macrophage phenotypes, and inflammatory cytokines were evaluated using immunohistochemistry techniques and enzyme-linked immunosorbent assays (ELISA) of the excised corneas. Compared to the PBS control group, MSC-exo treatment group had faster epithelial wound closure (0.041), lower corneal haze score (p = 0.002), and reduced haze intensity (p = 0.004) throughout the follow-up period. Attenuation of corneal vascularisation based on CD31 and LYVE-1 staining and reduced fibrosis as measured by fibronectin and collagen 3A1 staining was also observed in the MSC-exo group. MSC-exo treated corneas also displayed a regenerative immune phenotype characterized by a higher infiltration of CD163+, CD206+ M2 macrophages over CD80+, CD86+ M1 macrophages (p = 0.023), reduced levels of pro-inflammatory IL-1β, IL-8, and TNF-α, and increased levels of anti-inflammatory IL-10. In conclusion, topical MSC-exo could alleviate corneal insults by promoting wound closure and reducing scar development, possibly through anti-angiogenesis and immunomodulation towards a regenerative and anti-inflammatory phenotype.
Collapse
Affiliation(s)
- Hon Shing Ong
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Corneal and External Diseases Department, Singapore National Eye Centre, Singapore 168751, Singapore
| | - Andri K. Riau
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Gary Hin-Fai Yam
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Evelina J. Y. Han
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Tze-Wei Goh
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Ruenn Chai Lai
- Institute of Medical Biology & Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Sai Kiang Lim
- Institute of Medical Biology & Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Jodhbir S. Mehta
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Corneal and External Diseases Department, Singapore National Eye Centre, Singapore 168751, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
104
|
Pan W, Chen H, Wang A, Wang F, Zhang X. Challenges and strategies: Scalable and efficient production of mesenchymal stem cells-derived exosomes for cell-free therapy. Life Sci 2023; 319:121524. [PMID: 36828131 DOI: 10.1016/j.lfs.2023.121524] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/10/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023]
Abstract
Exosomes are small membrane vesicles secreted by most cell types, and widely exist in cell supernatants and various body fluids. They can transmit numerous bioactive elements, such as proteins, nucleic acids, and lipids, to affect the gene expression and function of recipient cells. Mesenchymal stem cells (MSCs) have been confirmed to be a potentially promising therapy for tissue repair and regeneration. Accumulating studies demonstrated that the predominant regenerative paradigm of MSCs transplantation was the paracrine effect but not the differentiation effect. Exosomes secreted by MSCs also showed similar therapeutic effects as their parent cells and were considered to be used for cell-free regenerative medicine. However, the inefficient and limited production has hampered their development for clinical translation. In this review, we summarize potential methods to efficiently promote the yield of exosomes. We mainly focus on engineering the process of exosome biogenesis and secretion, altering the cell culture conditions, cell expansion through 3D dynamic culture and the isolation of exosomes. In addition, we also discuss the application of MSCs-derived exosomes as therapeutics in disease treatment.
Collapse
Affiliation(s)
- Wei Pan
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongyuan Chen
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324 Jingwuweiqi Road 324, Jinan 250021, China
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, UC Davis Health Medical Center, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Fengshan Wang
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; National Glycoengineering Research Center, Shandong University, Jinan, Shandong 250012, China.
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
105
|
Dong J, Wu B, Tian W. Human adipose tissue-derived small extracellular vesicles promote soft tissue repair through modulating M1-to-M2 polarization of macrophages. Stem Cell Res Ther 2023; 14:67. [PMID: 37024970 PMCID: PMC10080905 DOI: 10.1186/s13287-023-03306-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Successful regenerative medicine strategies need the manipulation and control of macrophages' phenotypic switching. Our previous study indicated that rat and porcine adipose tissue-derived small extracellular vesicles could successfully promote soft tissue repair. However, whether human adipose tissue-derived small extracellular vesicles (h-sEV-AT) showed the same ability to promote soft tissue regeneration and whether adipose tissue-derived small extracellular vesicles (sEV-AT) contribute to modulating the polarization of macrophages were unknown. METHODS In this study, we, for the first time, isolated h-sEV-AT from liposuction adipose tissue and characterized the morphology, size distribution, and marker protein. In vitro, we treated adipose-derived stromal/stem cells (ASCs), endothelial cells (ECs), and M1 macrophages with h-sEV-AT. In vivo, the ability of h-sEV-AT to promote soft tissue regeneration and polarize macrophages was investigated. RESULTS The results indicated that h-sEV-AT possessed the characteristics of small extracellular vesicles (sEVs). In vitro, an obvious increase in adipogenesis and angiogenesis was induced by h-sEV-AT. In vivo, h-sEV-AT successfully induced the regeneration of adipose tissue and effectively accelerated full-thickness skin wound healing. Besides, we found that h-sEV-AT showed the ability to increase the percentage of M2 macrophages both in vivo and in vitro, which had been reported to contribute to tissue repair and regeneration. CONCLUSIONS Taken together, these results suggested that h-sEV-AT showed the ability to induce soft tissue repair supported by not only the differentiation of ASCs and ECs but also the polarization of macrophages. Considering the abundant sources, high yield, and guaranteed effectiveness, this study provided a cell-free strategy for soft tissue regeneration that directly isolated small extracellular vesicles from human liposuction adipose tissue.
Collapse
Affiliation(s)
- Jia Dong
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Sichuan, 610041, Chengdu, China.
- Department of Stomatology, People's Hospital of Longhua Shenzhen, Shenzhen, 518109, Guangdong, China.
| | - Bin Wu
- Department of Stomatology, People's Hospital of Longhua Shenzhen, Shenzhen, 518109, Guangdong, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Sichuan, 610041, Chengdu, China.
| |
Collapse
|
106
|
Spiers HVM, Stadler LKJ, Smith H, Kosmoliaptsis V. Extracellular Vesicles as Drug Delivery Systems in Organ Transplantation: The Next Frontier. Pharmaceutics 2023; 15:891. [PMID: 36986753 PMCID: PMC10052210 DOI: 10.3390/pharmaceutics15030891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Extracellular vesicles are lipid bilayer-delimited nanoparticles excreted into the extracellular space by all cells. They carry a cargo rich in proteins, lipids and DNA, as well as a full complement of RNA species, which they deliver to recipient cells to induce downstream signalling, and they play a key role in many physiological and pathological processes. There is evidence that native and hybrid EVs may be used as effective drug delivery systems, with their intrinsic ability to protect and deliver a functional cargo by utilising endogenous cellular mechanisms making them attractive as therapeutics. Organ transplantation is the gold standard for treatment for suitable patients with end-stage organ failure. However, significant challenges still remain in organ transplantation; prevention of graft rejection requires heavy immunosuppression and the lack of donor organs results in a failure to meet demand, as manifested by growing waiting lists. Pre-clinical studies have demonstrated the ability of EVs to prevent rejection in transplantation and mitigate ischemia reperfusion injury in several disease models. The findings of this work have made clinical translation of EVs possible, with several clinical trials actively recruiting patients. However, there is much to be uncovered, and it is essential to understand the mechanisms behind the therapeutic benefits of EVs. Machine perfusion of isolated organs provides an unparalleled platform for the investigation of EV biology and the testing of the pharmacokinetic and pharmacodynamic properties of EVs. This review classifies EVs and their biogenesis routes, and discusses the isolation and characterisation methods adopted by the international EV research community, before delving into what is known about EVs as drug delivery systems and why organ transplantation represents an ideal platform for their development as drug delivery systems.
Collapse
Affiliation(s)
- Harry V M Spiers
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Lukas K J Stadler
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Hugo Smith
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
107
|
Pang L, Jin H, Lu Z, Xie F, Shen H, Li X, Zhang X, Jiang X, Wu L, Zhang M, Zhang T, Zhai Y, Zhang Y, Guan H, Su J, Li M, Gao J. Treatment with Mesenchymal Stem Cell-Derived Nanovesicle-Containing Gelatin Methacryloyl Hydrogels Alleviates Osteoarthritis by Modulating Chondrogenesis and Macrophage Polarization. Adv Healthc Mater 2023:e2300315. [PMID: 36848378 DOI: 10.1002/adhm.202300315] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Indexed: 03/01/2023]
Abstract
Osteoarthritis is a degenerative disorder that can severely affect joints, and new treatment strategies are urgently needed. Administration of mesenchymal stem cell (MSC)-derived exosomes is a promising therapeutic strategy in osteoarthritis treatment. However, the poor yield of exosomes is an obstacle to the use of this modality in the clinic. Herein, a promising strategy is developed to fabricate high-yield exosome-mimicking MSC-derived nanovesicles (MSC-NVs) with enhanced regenerative and anti-inflammatory capabilities. MSC-NVs are prepared using an extrusion approach and are found to increase chondrocyte and human bone marrow MSC differentiation, proliferation, and migration, in addition to inducing M2 macrophage polarization. Furthermore, gelatin methacryloyl (GelMA) hydrogels loaded with MSC-NVs (GelMA-NVs) are formulated, which exhibit sustained release of MSC-NVs and are shown to be biocompatible with excellent mechanical properties. In a mouse osteoarthritis model constructed by surgical destabilization of the medial meniscus (DMM), GelMA-NVs effectively ameliorate osteoarthritis severity, reduce the secretion of catabolic factors, and enhance matrix synthesis. Furthermore, GelMA-NVs induce M2 macrophage polarization and inflammatory response inhibition in vivo. The findings demonstrate that GelMA-NVs hold promise for osteoarthritis treatment through modulation of chondrogenesis and macrophage polarization.
Collapse
Affiliation(s)
- Liying Pang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.,Department of Laboratory Medicine, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, 157011, China.,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Hong Jin
- Department of Laboratory Medicine, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Zhengmao Lu
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Huaxing Shen
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Xinying Li
- Department of Laboratory Medicine, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xinyi Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Xianghe Jiang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Mengya Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yonghua Zhai
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yuanyuan Zhang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Huilin Guan
- Department of Scientific Research, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.,Department of Orthopaedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200010, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
108
|
Gemayel J, Chaker D, El Hachem G, Mhanna M, Salemeh R, Hanna C, Harb F, Ibrahim A, Chebly A, Khalil C. Mesenchymal stem cells-derived secretome and extracellular vesicles: perspective and challenges in cancer therapy and clinical applications. Clin Transl Oncol 2023:10.1007/s12094-023-03115-7. [PMID: 36808392 DOI: 10.1007/s12094-023-03115-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/07/2023] [Indexed: 02/19/2023]
Abstract
Stem cell-based therapies have been foreshowed as a promising therapeutic approach for the treatment of several diseases. However, in the cancer context, results obtained from clinical studies were found to be quite limited. Deeply implicated in inflammatory cues, Mesenchymal, Neural, and Embryonic Stem Cells have mainly been used in clinical trials as a vehicle to deliver and stimulate signals in tumors niche. Although these stem cells have shown some therapeutical promises, they still face several challenges, including their isolation, immunosuppression potential, and tumorigenicity. In addition, regulatory and ethical concerns limit their use in several countries. Mesenchymal stem cells (MSC) have emerged as a gold standard adult stem cell medicine tool due to their distinctive characteristics, such as self-renewal and potency to differentiate into numerous cell types with lower ethical restrictions. Secreted extracellular vesicles (EVs), secretomes, and exosomes play a crucial role in mediating cell-to-cell communication to maintain physiological homeostasis and influence pathogenesis. Due to their low immunogenicity, biodegradability, low toxicity, and ability to transfer bioactive cargoes across biological barriers, EVs and exosomes were considered an alternative to stem cell therapy through their immunological features. MSCs-derived EVs, exosomes, and secretomes showed regenerative, anti-inflammatory, and immunomodulation properties while treating human diseases. In this review, we provide an overview of the paradigm of MSCs derived exosomes, secretome, and EVs cell-free-based therapies, we will focus on MSCs-derived components in anti-cancer treatment with decreased risk of immunogenicity and toxicity. Astute exploration of MSCs may lead to a new opportunity for efficient therapy for patients with cancer.
Collapse
Affiliation(s)
- Jack Gemayel
- Faculty of Health Sciences, Balamand University, Beirut, Lebanon
| | - Diana Chaker
- INSERM, National Institute of Health and Medical Research, Paris XI, Paris, France
- Reviva Stem Cell Platform for Research and Applications Center, Bsalim, Lebanon
| | - Georges El Hachem
- Balamand University, Faculty of Medicine, Beirut, Lebanon
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Al Kurah, P.O. Box 100, Kalhat, Lebanon
| | - Melissa Mhanna
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Rawad Salemeh
- Reviva Stem Cell Platform for Research and Applications Center, Bsalim, Lebanon
| | - Colette Hanna
- Faculty of Medicine, Lebanese American University Medical Center, Rizk Hospital, Beirut, Lebanon
| | - Frederic Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Al Kurah, P.O. Box 100, Kalhat, Lebanon
| | - Ahmad Ibrahim
- Reviva Stem Cell Platform for Research and Applications Center, Bsalim, Lebanon
- Balamand University, Faculty of Medicine, Beirut, Lebanon
| | - Alain Chebly
- Medical Genetics Unit, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
- Higher Institute of Public Health, Saint Joseph University, Beirut, Lebanon
| | - Charbel Khalil
- Reviva Stem Cell Platform for Research and Applications Center, Bsalim, Lebanon.
- Bone Marrow Transplant Unit, Burjeel Medical City, Abu Dhabi, UAE.
- Stem Cell Institute, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
109
|
Li X, Duan H, Wang S, Lv CX. Umbilical cord mesenchymal stem cell-derived exosomes reverse endometrial fibrosis by the miR-145-5p/ZEB2 axis in intrauterine adhesions. Reprod Biomed Online 2023; 46:234-243. [PMID: 36567149 DOI: 10.1016/j.rbmo.2022.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/21/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
RESEARCH QUESTION What is the specific mechanism of umbilical cord mesenchymal stem cell-derived exosomes (UCMSC-exos) in regulating endometrial repair and regeneration? DESIGN In this study, UCMSC-exos were harvested by differential ultracentrifugation from umbilical cord mesenchymal stem cell culture supernatant and identified with western blotting, transmission electron microscopy and nanoparticle tracking analysis. Transforming growth factor-β1 (TGFβ1) at different concentrations was used to construct the intrauterine adhesions cell model. The fibrotic markers were assessed by quantitative reverse transcription-polymerase chain reaction and western blotting. The effects of miR-145-5p over-expression on endometrial fibrosis were assessed. Dual luciferase assay was performed to verify the relationship between miR-145-5p and zinc finger E-box binding homeobox 2 (ZEB2). RESULTS The isolated UCMSC-exos had a typical cup-shaped morphology, expressed the specific exosomal markers Alix, CD63 and TSG101, and were approximately 50-150 nm in diameter. TGFβ1 at 10 ng/ml significantly promoted endometrial fibrosis, which was reversed by 20 µg/ml UCMSC-exos. Exosomal miR-145-5p ameliorated TGFβ1-induced endometrial fibrosis. ZEB2 was inversely regulated by exosomal miR-145-5p as a direct target. CONCLUSIONS UCMSC-exos might reverse endometrial stromal cell fibrosis by regulating the miR-145-5p/ZEB2 axis, representing a potential novel strategy to promote endometrial repair.
Collapse
Affiliation(s)
- Xiao Li
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Hua Duan
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China.
| | - Sha Wang
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Cheng-Xiao Lv
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| |
Collapse
|
110
|
Jenner F, Wagner A, Gerner I, Ludewig E, Trujanovic R, Rohde E, von Rechenberg B, Gimona M, Traweger A. Evaluation of the Potential of Umbilical Cord Mesenchymal Stromal Cell-Derived Small Extracellular Vesicles to Improve Rotator Cuff Healing: A Pilot Ovine Study. Am J Sports Med 2023; 51:331-342. [PMID: 36645050 DOI: 10.1177/03635465221145958] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Despite significant advancements in surgical techniques to repair rotator cuff (RC) injuries, failure rates remain high and novel approaches to adequately overcome the natural biological limits of tendon and enthesis regeneration of the RC are required. Small extracellular vesicles (sEVs) derived from the secretome of human multipotent mesenchymal stromal cells (MSCs) have been demonstrated to modulate inflammation and reduce fibrotic adhesions, and therefore their local application could improve outcomes after RC repair. PURPOSE In this pilot study, we evaluated the efficacy of clinical-grade human umbilical cord (hUC) MSC-derived sEVs (hUC-MSC-sEVs) loaded onto a type 1 collagen scaffold in an ovine model of acute infraspinatus tendon injury to improve RC healing. STUDY DESIGN Controlled laboratory study. METHODS sEVs were enriched from hUC-MSC culture media and were characterized by surface marker profiling. The immunomodulatory capacity was evaluated in vitro by T-cell proliferation assays, and particle count was determined by nanoparticle tracking analysis. Twelve skeletally mature sheep were subjected to partial infraspinatus tenotomy and enthesis debridement. The defects of 6 animals were treated with 2 × 1010 hUC-MSC-sEVs loaded onto a type 1 collagen sponge, whereas 6 animals received only a collagen sponge, serving as controls. Six weeks postoperatively, the healing of the infraspinatus tendon and the enthesis was evaluated by magnetic resonance imaging (MRI) and hard tissue histology. RESULTS CD3/CD28-stimulated T-cell proliferation was significantly inhibited by hUC-MSC-sEVs (P = .015) that displayed the typical surface marker profile, including the presence of the MSC marker proteins CD44 and melanoma-associated chondroitin sulfate proteoglycan. The local application of hUC-MSC-sEVs did not result in any marked systemic adverse events. Histologically, significantly improved Watkins scores (P = .031) indicated improved tendon and tendon-to-bone insertion repair after sEV treatment and lower postcontrast signal of the tendon and adjacent structures on MRI suggested less residual inflammation at the defect area. Furthermore, the formation of osteophytes at the injury site was significantly attenuated (P = .037). CONCLUSION A local, single-dose application of hUC-MSC-sEVs promoted tendon and enthesis healing in an ovine model of acute RC injury. CLINICAL RELEVANCE Surgical repair of RC tears generally results in a clinical benefit for the patient; however, considerable rerupture rates have been reported. sEVs have potential as a cell-free biotherapeutic to improve healing outcomes after RC injury.
Collapse
Affiliation(s)
- Florien Jenner
- VETERM, Equine Surgery Unit, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andrea Wagner
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University, Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Iris Gerner
- VETERM, Equine Surgery Unit, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Eberhard Ludewig
- Diagnostic Imaging Unit, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Robert Trujanovic
- Clinical Unit of Anaesthesiology and Perioperative Intensive Care, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eva Rohde
- Department of Transfusion Medicine, Salzburger Landeskliniken GesmbH, Paracelsus Medical University, Salzburg, Austria.,GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University, Salzburg, Austria.,Research Program "Nanovesicular Therapies," Paracelsus Medical University, Salzburg, Austria
| | - Andreas Traweger
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University, Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
111
|
Exploring the role of exosomes in rheumatoid arthritis. Inflammopharmacology 2023; 31:119-128. [PMID: 36414831 DOI: 10.1007/s10787-022-01100-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022]
Abstract
In prosperous countries, autoimmune illnesses affect minimum 7% of the community. Rheumatoid Arthritis (RA) as an autoimmune illness is thought to be induced through a variety of genomic, physiological, and biological factors. Many experts in the field of nanomedicine have looked to stem cells as a viable strategy to repair human tissue; however, exosomes have demonstrated greater potential in recent years. Exosomes, produced from stem cells in particular, have exhibited a high propensity to give therapeutic effects. To resist local cellular stress, they are secreted in a paracrine manner from cells. As a result, exosomes produced from stem cells can provide enormous health uses. If treatment is not given, autoantibodies produce synovial inflammation and arthritis, which can lead to chronic inflammation, and impairment. Exosomes could be administered for the treatment of RA, by acting as therapeutic vectors. Exosomes are murine extracellular vesicles that influence biological mechanisms and signal transduction by transporting genetic and protein components. Diseases like RA and bone fractures could be treated using cell-free therapeutic strategies if exosomes could be isolated from stem cells efficiently and packaged with specific restorative substances. To get to this position, many breakthroughs must be achieved, and the following review summarises the most recent developments in stem cell-derived exosomes, with a focus on the important literature on exosome dynamics in RA.
Collapse
|
112
|
Dai Y, Chen Y, Hu Y, Zhang L. Current knowledge and future perspectives on exosomes in the field of regenerative medicine: a bibliometric analysis. Regen Med 2023; 18:123-136. [PMID: 36325823 DOI: 10.2217/rme-2022-0141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective: This study aimed to use bibliometric analysis to qualitatively and quantitatively evaluate the research of exosomes in the field of regenerative medicine and to provide research hotspots and trends in this field. Materials & methods: Bibliometric analysis and data presentation were performed by VOSviewer and Microsoft Excel. Results: China was the major contributor to research in this field and enjoys a high reputation in academia. The highest contributing institution is Shanghai Jiao Tong University. Research hotspots included exosome-mediated neurovascular regeneration, exosome mechanism research, exosome-mediated cartilage regeneration and repair and exosome-mediated cardiac regeneration. Research was trending in the treatment of osteoarthritis, knee disease and cartilage regeneration and repair. Conclusion: This study provides a panoramic view of the application of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Yuxuan Dai
- Department of Plastic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Yu Chen
- Division of Thyroid Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yiming Hu
- Department of Plastic & Aesthetic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Lianbo Zhang
- Department of Plastic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| |
Collapse
|
113
|
Sanz-Ros J, Mas-Bargues C, Romero-García N, Huete-Acevedo J, Dromant M, Borrás C. Extracellular Vesicles as Therapeutic Resources in the Clinical Environment. Int J Mol Sci 2023; 24:2344. [PMID: 36768664 PMCID: PMC9917082 DOI: 10.3390/ijms24032344] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
The native role of extracellular vesicles (EVs) in mediating the transfer of biomolecules between cells has raised the possibility to use them as therapeutic vehicles. The development of therapies based on EVs is now expanding rapidly; here we will describe the current knowledge on different key points regarding the use of EVs in a clinical setting. These points are related to cell sources of EVs, isolation, storage, and delivery methods, as well as modifications to the releasing cells for improved production of EVs. Finally, we will depict the application of EVs therapies in clinical trials, considering the impact of the COVID-19 pandemic on the development of these therapies, pointing out that although it is a promising therapy for human diseases, we are still in the initial phase of its application to patients.
Collapse
Affiliation(s)
- Jorge Sanz-Ros
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Department of Cardiology, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Nekane Romero-García
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Department of Anesthesiology and Surgical Trauma Intensive Care, Hospital Clinic Universitari de Valencia, University of Valencia, 46010 Valencia, Spain
| | - Javier Huete-Acevedo
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Mar Dromant
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
114
|
Oveili E, Vafaei S, Bazavar H, Eslami Y, Mamaghanizadeh E, Yasamineh S, Gholizadeh O. The potential use of mesenchymal stem cells-derived exosomes as microRNAs delivery systems in different diseases. Cell Commun Signal 2023; 21:20. [PMID: 36690996 PMCID: PMC9869323 DOI: 10.1186/s12964-022-01017-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/14/2022] [Indexed: 01/24/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of small non-coding RNAs that regulate gene expression by targeting mRNA. Moreover, it has been shown that miRNAs expression are changed in various diseases, such as cancers, autoimmune disease, infectious diseases, and neurodegenerative Diseases. The suppression of miRNA function can be easily attained by utilizing of anti-miRNAs. In contrast, an enhancement in miRNA function can be achieved through the utilization of modified miRNA mimetics. The discovery of appropriate miRNA carriers in the body has become an interesting subject for investigators. Exosomes (EXOs) therapeutic efficiency and safety for transferring different cellular biological components to the recipient cell have attracted significant attention for their capability as miRNA carriers. Mesenchymal stem cells (MSCs) are recognized to generate a wide range of EXOs (MSC-EXOs), showing that MSCs may be effective for EXO generation in a clinically appropriate measure as compared to other cell origins. MSC-EXOs have been widely investigated because of their immune attributes, tumor-homing attributes, and flexible characteristics. In this article, we summarized the features of miRNAs and MSC-EXOs, including production, purification, and miRNA loading methods of MSC-EXOs, and the modification of MSC-EXOs for targeted miRNA delivery in various diseases. Video abstract.
Collapse
Affiliation(s)
- Elham Oveili
- Department of Pharmaceutical Science, Azad Islamic University of Medical Sciences, Tehran, Iran
| | - Somayeh Vafaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Bazavar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yeganeh Eslami
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ehsan Mamaghanizadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saman Yasamineh
- Department of Biotechnology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Omid Gholizadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
115
|
Liu F, Sun T, An Y, Ming L, Li Y, Zhou Z, Shang F. The potential therapeutic role of extracellular vesicles in critical-size bone defects: Spring of cell-free regenerative medicine is coming. Front Bioeng Biotechnol 2023; 11:1050916. [PMID: 36733961 PMCID: PMC9887316 DOI: 10.3389/fbioe.2023.1050916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
In recent years, the incidence of critical-size bone defects has significantly increased. Critical-size bone defects seriously affect patients' motor functions and quality of life and increase the need for additional clinical treatments. Bone tissue engineering (BTE) has made great progress in repairing critical-size bone defects. As one of the main components of bone tissue engineering, stem cell-based therapy is considered a potential effective strategy to regenerate bone tissues. However, there are some disadvantages including phenotypic changes, immune rejection, potential tumorigenicity, low homing efficiency and cell survival rate that restrict its wider clinical applications. Evidence has shown that the positive biological effects of stem cells on tissue repair are largely mediated through paracrine action by nanostructured extracellular vesicles (EVs), which may overcome the limitations of traditional stem cell-based treatments. In addition to stem cell-derived extracellular vesicles, the potential therapeutic roles of nonstem cell-derived extracellular vesicles in critical-size bone defect repair have also attracted attention from scholars in recent years. Currently, the development of extracellular vesicles-mediated cell-free regenerative medicine is still in the preliminary stage, and the specific mechanisms remain elusive. Herein, the authors first review the research progress and possible mechanisms of extracellular vesicles combined with bone tissue engineering scaffolds to promote bone regeneration via bioactive molecules. Engineering modified extracellular vesicles is an emerging component of bone tissue engineering and its main progression and clinical applications will be discussed. Finally, future perspectives and challenges of developing extracellular vesicle-based regenerative medicine will be given. This review may provide a theoretical basis for the future development of extracellular vesicle-based biomedicine and provide clinical references for promoting the repair of critical-size bone defects.
Collapse
Affiliation(s)
- Fen Liu
- Department of Periodontology, Shenzhen Stomatological Hospital (Pingshan), Southern Medical University, Shenzhen, Guangdong, China
| | - Tianyu Sun
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying An
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture and Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Leiguo Ming
- Department of Research and Development, Shaanxi Zhonghong Institute of Regenerative Medicine, Xi’an, Shaanxi, China
| | - Yinghui Li
- Department of Orthodontics, Stomatological Hospital, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, Tibet, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| | - Fengqing Shang
- Department of Stomatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| |
Collapse
|
116
|
Wang H, Sun XC, Li JH, Yin LQ, Yan YF, Ma X, Xia HF. Combining Bone Collagen Material with hUC-MSCs for Applicationto Spina Bifida in a Rabbit Model. Stem Cell Rev Rep 2023; 19:1034-1050. [PMID: 36648605 DOI: 10.1007/s12015-022-10478-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 01/18/2023]
Abstract
Spina bifida is one of the neural tube defects, with a high incidence in human birth defects, which seriously affects the health and quality of life of patients. In the treatment of bone defects, the source of autologous bone is limited and will cause secondary damage to the patient. At the same time, since the bone tissue in animals needs to play a variety of biological functions, its complex structure cannot be replaced by a single material. The combination of mechanical materials and biological materials has become a common choice. Human umbilical cord mesenchymal stem cells (hUC-MSCs) have the advantages of easy access, rapid proliferation, low immunogenicity, and no ethical issues. It is often used in the clinical research of tissue regeneration and repair. Therefore, in this study, we established a spina bifida model using Japanese white rabbits. This model was used to screen the best regenerative repair products for congenital spina bifida, and to evaluate the safety of regenerative repair products. The results showed that the combination of hUC-MSCs with collagen material had better regeneration effect than collagen material alone, and had no negative impact on the health of animals. This study provides a new idea for the clinical treatment of spina bifida, and also helps to speed up the research progress of regenerative repair products.
Collapse
Affiliation(s)
- Hu Wang
- Reproductive and Genetic Center, NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China.,Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xue-Cheng Sun
- Reproductive and Genetic Center, NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China.,Medical Genetics, Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Jian-Hui Li
- Reproductive and Genetic Center, NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China
| | - Li-Qiang Yin
- Yantai Zhenghai Bio-Tech Co., Ltd., Yantai, 264006, China
| | - Yu-Fang Yan
- Yantai Zhenghai Bio-Tech Co., Ltd., Yantai, 264006, China
| | - Xu Ma
- Reproductive and Genetic Center, NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China. .,Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Hong-Fei Xia
- Reproductive and Genetic Center, NHC Key Laboratory of Reproductive Health Engineering Technology Research (NRIFP), National Research Institute for Family Planning, Beijing, 100081, China. .,Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
117
|
Rao DY, Huang DF, Si MY, Lu H, Tang ZX, Zhang ZX. Role of exosomes in non-small cell lung cancer and EGFR-mutated lung cancer. Front Immunol 2023; 14:1142539. [PMID: 37122754 PMCID: PMC10130367 DOI: 10.3389/fimmu.2023.1142539] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/22/2023] [Indexed: 05/02/2023] Open
Abstract
As an important mediator of information transfer between cells, exosomes play a unique role in regulating tumor growth, supporting vascular proliferation, tumor invasion, and metastasis. Exosomes are widely present in various body fluids, and therefore they can be used as a potential tool for non-invasive liquid biopsy. The present study reviews the role of exosomes in liquid biopsy, tumor microenvironment formation, and epithelial-mesenchymal transition in non-small cell lung cancer (NSCLC). By targeting epidermal growth factor receptor (EGFR) therapy as a first-line treatment for patients with NSCLC, this study also briefly describes the occurrence of EGRF+ exosomes and the role of exosomes and their contents in non-invasive detection and potential therapeutic targets in EGFR-mutated lung cancer.
Collapse
Affiliation(s)
- Ding-Yu Rao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - De-Fa Huang
- Laboratory Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mao-Yan Si
- The First Clinical College, Gannan Medical University, Ganzhou, China
| | - Hua Lu
- The First Clinical College, Southern Medical University, Guangzhou, China
| | - Zhi-Xian Tang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Zhi-Xian Tang, ; Zu-Xiong Zhang,
| | - Zu-Xiong Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
118
|
Chin AR. Challenges for Studying and Isolating Extracellular Vesicles from Cell-Conditioned Media. Methods Mol Biol 2023; 2666:299-315. [PMID: 37166673 DOI: 10.1007/978-1-0716-3191-1_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Extracellular vesicles (EV) are small (100-1000 nm) particles that cells release into the extracellular space that have become increasingly famous for their potential in regenerative medicine and for their alterations in diseases such as cancer to promote disease progression, in particular for their potential for intercellular communication. However, studying EV can be challenging due to the broad diversity of both the EV themselves as well as the methods used to study them. This chapter aims to help investigators new to the EV field by describing challenges with studying EV, methods for enriching EV, and a simple EV enrichment protocol using differential ultracentrifugation.
Collapse
Affiliation(s)
- Andrew R Chin
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, USA.
| |
Collapse
|
119
|
Ganesh V, Seol D, Gomez-Contreras PC, Keen HL, Shin K, Martin JA. Exosome-Based Cell Homing and Angiogenic Differentiation for Dental Pulp Regeneration. Int J Mol Sci 2022; 24:466. [PMID: 36613910 PMCID: PMC9820194 DOI: 10.3390/ijms24010466] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/21/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Exosomes have attracted attention due to their ability to promote intercellular communication leading to enhanced cell recruitment, lineage-specific differentiation, and tissue regeneration. The object of this study was to determine the effect of exosomes on cell homing and angiogenic differentiation for pulp regeneration. Exosomes (DPSC-Exos) were isolated from rabbit dental pulp stem cells cultured under a growth (Exo-G) or angiogenic differentiation (Exo-A) condition. The characterization of exosomes was confirmed by nanoparticle tracking analysis and an antibody array. DPSC-Exos significantly promoted cell proliferation and migration when treated with 5 × 108/mL exosomes. In gene expression analysis, DPSC-Exos enhanced the expression of angiogenic markers including vascular endothelial growth factor A (VEGFA), Fms-related tyrosine kinase 1 (FLT1), and platelet and endothelial cell adhesion molecule 1 (PECAM1). Moreover, we identified key exosomal microRNAs in Exo-A for cell homing and angiogenesis. In conclusion, the exosome-based cell homing and angiogenic differentiation strategy has significant therapeutic potential for pulp regeneration.
Collapse
Affiliation(s)
- Venkateswaran Ganesh
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
- Department of Roy J. Carver Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Dongrim Seol
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
- Department of Orthodontics, University of Iowa, Iowa City, IA 52242, USA
| | | | - Henry L. Keen
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Kyungsup Shin
- Department of Orthodontics, University of Iowa, Iowa City, IA 52242, USA
| | - James A. Martin
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
- Department of Roy J. Carver Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
120
|
Mélou C, Pellen-Mussi P, Jeanne S, Novella A, Tricot-Doleux S, Chauvel-Lebret D. Osteoarthritis of the Temporomandibular Joint: A Narrative Overview. MEDICINA (KAUNAS, LITHUANIA) 2022; 59:medicina59010008. [PMID: 36676632 PMCID: PMC9866170 DOI: 10.3390/medicina59010008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Background and Objectives: This study reviewed the literature to summarize the current and recent knowledge of temporomandibular joint osteoarthritis (TMJOA). Methods: Through a literature review, this work summarizes many concepts related to TMJOA. Results: Although many signaling pathways have been investigated, the etiopathogenesis of TMJOA remains unclear. Some clinical signs are suggestive of TMJOA; however, diagnosis is mainly based on radiological findings. Treatment options include noninvasive, minimally invasive, and surgical techniques. Several study models have been used in TMJOA studies because there is no gold standard model. Conclusion: More research is needed to develop curative treatments for TMJOA, which could be tested with reliable in vitro models, and to explore tissue engineering to regenerate damaged temporomandibular joints.
Collapse
Affiliation(s)
- Caroline Mélou
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University Rennes, UMR 6226, 35000 Rennes, France
- CHU Rennes, Pôle d’Odontologie, 35033 Rennes, France
- UFR Odontologie, 35043 Rennes, France
| | - Pascal Pellen-Mussi
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University Rennes, UMR 6226, 35000 Rennes, France
| | - Sylvie Jeanne
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University Rennes, UMR 6226, 35000 Rennes, France
- CHU Rennes, Pôle d’Odontologie, 35033 Rennes, France
- UFR Odontologie, 35043 Rennes, France
| | - Agnès Novella
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University Rennes, UMR 6226, 35000 Rennes, France
| | - Sylvie Tricot-Doleux
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University Rennes, UMR 6226, 35000 Rennes, France
| | - Dominique Chauvel-Lebret
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University Rennes, UMR 6226, 35000 Rennes, France
- CHU Rennes, Pôle d’Odontologie, 35033 Rennes, France
- UFR Odontologie, 35043 Rennes, France
- Correspondence: ; Tel.: +33-2-23-23-43-64; Fax: +33-2-23-23-43-93
| |
Collapse
|
121
|
Shi Y, Shao J, Zhang Z, Zhang J, Lu H. Effect of condylar chondrocyte exosomes on condylar cartilage osteogenesis in rats under tensile stress. Front Bioeng Biotechnol 2022; 10:1061855. [PMID: 36561044 PMCID: PMC9766957 DOI: 10.3389/fbioe.2022.1061855] [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: 10/05/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Functional orthoses are commonly used to treat skeletal Class II malocclusion, but the specific mechanism through which they do this has been a challenging topic in orthodontics. In the present study, we aimed to explore the effect of tensile stress on the osteogenic differentiation of condylar chondrocytes from an exosomal perspective. Methods: We cultured rat condylar chondrocytes under resting and tensile stress conditions and subsequently extracted cellular exosomes from them. We then screened miRNAs that were differentially expressed between the two exosome extracts by high-throughput sequencing and performed bioinformatics analysis and osteogenesis-related target gene prediction using the TargetScan and miRanda softwares. Exosomes cultured under resting and tensile stress conditions were co-cultured with condylar chondrocytes for 24 h to form the Control-Exo and Force-Exo exosome groups, respectively. Quantitative real time PCR(RT-qPCR) and western blotting were then used to determine the mRNA and protein expression levels of Runx2 and Sox9 in condylar chondrocytes. Results: The mRNA and protein expression levels of Runx2 and Sox9 in the Force-Exo group were significantly higher than those in the Control-Exo group (p < 0.05). The differential miRNA expression results were consistent with our sequencing results. Bioinformatics analysis and target gene prediction results showed that the main biological processes and molecular functions involved in differential miRNA expression in exosomes under tensile stress were biological processes and protein binding, respectively. Kyoto Gene and Genome Data Bank (KEGG) pathway enrichment analysis showed significant enrichment of differentially expressed miRNAs in the mTOR signaling pathway. The differentially expressed miRNAs were found to target osteogenesis-related genes. Conclusion: These results suggest that stimulation of rat condylar chondrocytes with tensile stress can alter the expression levels of certain miRNAs in their exosomes and promote their osteogenic differentiation. Exosomes under tensile stress culture conditions thus have potential applications in the treatment of Osteoarthritis (OA).
Collapse
Affiliation(s)
- Yuan Shi
- Department of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Shao
- Department of Stomatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Zanzan Zhang
- Department of Stomatology, Ningbo No. 2 Hospital, Ningbo, China
| | - Jianan Zhang
- Department of Dentistry, Center of Orthodontics, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Haiping Lu
- Department of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China,*Correspondence: Haiping Lu,
| |
Collapse
|
122
|
Dong J, Wu B, Tian W. Adipose tissue-derived small extracellular vesicles modulate macrophages to improve the homing of adipocyte precursors and endothelial cells in adipose tissue regeneration. Front Cell Dev Biol 2022; 10:1075233. [PMID: 36561367 PMCID: PMC9763459 DOI: 10.3389/fcell.2022.1075233] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Rapid infiltration of endogenous cells induced by cell-free biomaterials is the first and crucial step in tissue regeneration and macrophage is largely involved. Our previous study reported adipose tissue-derived small extracellular vesicles (sEV-AT) could successfully promote adipose tissue regeneration. However, the role of macrophages in this process was unknown. In this study, we isolated sEV-AT and subcutaneously implanted it into the back of SD rats. The results showed sEV-AT increased macrophage infiltration significantly, which was followed by improving homing of adipocyte precursors (APs) and endothelial cells (ECs). However, when macrophages were depleted by clodronate liposome within 1 week, the homing of APs and ECs, and adipose tissue regeneration were destroyed. In vitro, sEV-AT showed the ability to promote the migration of macrophages directly. Besides, sEV-AT-pretreated macrophages improved the migration of APs and ECs, accompanied by the increase of chemokines (MCP-1, SDF-1, VEGF, and FGF) and the activation of NF-kB signaling pathway. These findings indicated sEV-AT might regulate the secretion of chemokines via activating NF-kB signaling pathway to improve homing of APs and ECs and facilitate adipose tissue regeneration. These findings deepened our understanding of small extracellular vesicle-induced tissue regeneration and laid a theoretical foundation for the clinical application of sEV-AT.
Collapse
Affiliation(s)
- Jia Dong
- Department of Stomatology, The People’s Hospital of Longhua Shenzhen, Shenzhen, China
| | - Bin Wu
- Department of Stomatology, The People’s Hospital of Longhua Shenzhen, Shenzhen, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease, National Engineering Laboratory for Oral Regenerative Medicine, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China,*Correspondence: Weidong Tian,
| |
Collapse
|
123
|
Xu XL, Xue Y, Ding JY, Zhu ZH, Wu XC, Song YJ, Cao YL, Tang LG, Ding DF, Xu JG. Nanodevices for deep cartilage penetration. Acta Biomater 2022; 154:23-48. [PMID: 36243371 DOI: 10.1016/j.actbio.2022.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease and is the main cause of chronic pain and functional disability in adults. Articular cartilage is a hydrated soft tissue that is composed of normally quiescent chondrocytes at a low density, a dense network of collagen fibrils with a pore size of 60-200 nm, and aggrecan proteoglycans with high-density negative charge. Although certain drugs, nucleic acids, and proteins have the potential to slow the progression of OA and restore the joints, these treatments have not been clinically applied owing to the lack of an effective delivery system capable of breaking through the cartilage barrier. Recently, the development of nanotechnology for delivery systems renders new ideas and treatment methods viable in overcoming the limited penetration. In this review, we focus on current research on such applications of nanotechnology, including exosomes, protein-based cationic nanocarriers, cationic liposomes/solid lipid nanoparticles, amino acid-based nanocarriers, polyamide derivatives-based nanocarriers, manganese dioxide, and carbon nanotubes. Exosomes are the smallest known nanoscale extracellular vesicles, and they can quickly deliver nucleic acids or proteins to the required depth. Through electrostatic interactions, nanocarriers with appropriate balance in cationic property and particle size have a strong ability to penetrate cartilage. Although substantial preclinical evidence has been obtained, further optimization is necessary for clinical transformation. STATEMENT OF SIGNIFICANCE: The dense cartilage matrix with high-negative charge was associated with reduced therapeutic effect in osteoarthritis patients with deep pathological changes. However, a systematic review in nanodevices for deep cartilage penetration is still lacking. Current approaches to assure penetration of nanosystems into the depth of cartilage were reviewed, including nanoscale extracellular vesicles from different cell lines and nanocarriers with appropriate balance in cationic property and size particle. Moreover, nanodevices entering clinical trials and further optimization were also discussed, providing important guiding significance to future research.
Collapse
Affiliation(s)
- Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yan Xue
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Centre), School of Medicine, Tongji University, Shanghai 201613, China
| | - Jia-Ying Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhi-Heng Zhu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi-Chen Wu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yong-Jia Song
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue-Long Cao
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Long-Guang Tang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China.
| | - Dao-Fang Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jian-Guang Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
124
|
Zelinka A, Roelofs AJ, Kandel RA, De Bari C. Cellular therapy and tissue engineering for cartilage repair. Osteoarthritis Cartilage 2022; 30:1547-1560. [PMID: 36150678 DOI: 10.1016/j.joca.2022.07.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/02/2023]
Abstract
Articular cartilage (AC) has limited capacity for repair. The first attempt to repair cartilage using tissue engineering was reported in 1977. Since then, cell-based interventions have entered clinical practice in orthopaedics, and several tissue engineering approaches to repair cartilage are in the translational pipeline towards clinical application. Classically, these involve a scaffold, substrate or matrix to provide structure, and cells such as chondrocytes or mesenchymal stromal cells to generate the tissue. We discuss the advantages and drawbacks of the use of various cell types, natural and synthetic scaffolds, multiphasic or gradient-based scaffolds, and self-organizing or self-assembling scaffold-free systems, for the engineering of cartilage constructs. Several challenges persist including achieving zonal tissue organization and integration with the surrounding tissue upon implantation. Approaches to improve cartilage thickness, organization and mechanical properties include mechanical stimulation, culture under hypoxic conditions, and stimulation with growth factors or other macromolecules. In addition, advanced technologies such as bioreactors, biosensors and 3D bioprinting are actively being explored. Understanding the underlying mechanisms of action of cell therapy and tissue engineering approaches will help improve and refine therapy development. Finally, we discuss recent studies of the intrinsic cellular and molecular mechanisms of cartilage repair that have identified novel signals and targets and are inspiring the development of molecular therapies to enhance the recruitment and cartilage reparative activity of joint-resident stem and progenitor cells. A one-fits-all solution is unrealistic, and identifying patients who will respond to a specific targeted treatment will be critical.
Collapse
Affiliation(s)
- A Zelinka
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Dept. Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - A J Roelofs
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - R A Kandel
- Lunenfeld Tanenbaum Research Institute, Sinai Health, Dept. Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| | - C De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
125
|
Liu Z, Wang S, Huo N, Yang S, Shi Q, Xu J. Extracellular vesicles: A potential future strategy for dental and maxillofacial tissue repair and regeneration. Front Physiol 2022; 13:1012241. [PMID: 36479350 PMCID: PMC9719951 DOI: 10.3389/fphys.2022.1012241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/09/2022] [Indexed: 06/18/2024] Open
Abstract
Extracellular vesicles (EVs), nano-sized bilayer membrane structures containing lipids, proteins and nucleic acids, play key roles in intercellular communication. Compared to stem cells, EVs have lower tumorigenicity and immunogenicity, are easier to manage and cause fewer ethic problems. In recent years, EVs have emerged as a potential solution for tissue regeneration in stomatology through cell-free therapies. The present review focuses on the role of EVs in dental and maxillofacial tissue repair and regeneration, including in dental and periodontal tissue, maxilla and mandible bone, temporomandibular joint cartilage, peripheral nerve and soft tissue. We also make a brief overview on the mechanism of EVs performing functions. However, limitations and challenges in clinical application of EVs still exist and should be addressed in future researches.
Collapse
Affiliation(s)
- Ziwei Liu
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Orthopedic Laboratory of PLA General Hospital, Beijing, China
| | - Situo Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Orthopedic Laboratory of PLA General Hospital, Beijing, China
| | - Na Huo
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuo Yang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Quan Shi
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Juan Xu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
126
|
Xiong Y, Mi BB, Lin Z, Hu YQ, Yu L, Zha KK, Panayi AC, Yu T, Chen L, Liu ZP, Patel A, Feng Q, Zhou SH, Liu GH. The role of the immune microenvironment in bone, cartilage, and soft tissue regeneration: from mechanism to therapeutic opportunity. Mil Med Res 2022; 9:65. [PMID: 36401295 PMCID: PMC9675067 DOI: 10.1186/s40779-022-00426-8] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Bone, cartilage, and soft tissue regeneration is a complex spatiotemporal process recruiting a variety of cell types, whose activity and interplay must be precisely mediated for effective healing post-injury. Although extensive strides have been made in the understanding of the immune microenvironment processes governing bone, cartilage, and soft tissue regeneration, effective clinical translation of these mechanisms remains a challenge. Regulation of the immune microenvironment is increasingly becoming a favorable target for bone, cartilage, and soft tissue regeneration; therefore, an in-depth understanding of the communication between immune cells and functional tissue cells would be valuable. Herein, we review the regulatory role of the immune microenvironment in the promotion and maintenance of stem cell states in the context of bone, cartilage, and soft tissue repair and regeneration. We discuss the roles of various immune cell subsets in bone, cartilage, and soft tissue repair and regeneration processes and introduce novel strategies, for example, biomaterial-targeting of immune cell activity, aimed at regulating healing. Understanding the mechanisms of the crosstalk between the immune microenvironment and regeneration pathways may shed light on new therapeutic opportunities for enhancing bone, cartilage, and soft tissue regeneration through regulation of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Bo-Bin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yi-Qiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Kang-Kang Zha
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany
| | - Zhen-Ping Liu
- Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany.,Joint Laboratory of Optofluidic Technology and System,National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, China
| | - Anish Patel
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China.
| | - Shuan-Hu Zhou
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
127
|
Esmaeili A, Hosseini S, Kamali A, Hosseinzadeh M, Shekari F, Baghaban Eslaminejad M. Co-aggregation of MSC/chondrocyte in a dynamic 3D culture elevates the therapeutic effect of secreted extracellular vesicles on osteoarthritis in a rat model. Sci Rep 2022; 12:19827. [PMID: 36400827 PMCID: PMC9674636 DOI: 10.1038/s41598-022-22592-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular vesicles (EVs) have therapeutic effects on osteoarthritis (OA). Some recent strategies could elevate EV's therapeutic properties including cell aggregation, co-culture, and 3D culture. It seems that a combination of these strategies could augment EV production and therapeutic potential. The current study aims to evaluate the quantity of EV yield and the therapeutic effect of EVs harvested from rabbit mesenchymal stem cells (MSCs) aggregates, chondrocyte aggregates, and their co-aggregates in a dynamic 3D culture in a rat osteoarthritis model. MSC and chondrocytes were aggregated and co-aggregated by spinner flasks, and their conditioned medium was collected. EVs were isolated by size exclusion chromatography and characterized in terms of size, morphology and surface markers. The chondrogenic potential of the MSC-ag, Cho-ag and Co-ag EVs on MSC micromass differentiation in chondrogenic media were assessed by qRT-PCR, histological and immunohistochemical analysis. 50 μg of MSC-ag-EVs, Cho-ag-EVs and Co-ag-EVs was injected intra-articularly per knee of OA models established by monoiodoacetate in rats. After 8 weeks follow up, the knee joints were harvested and analyzed by radiographic, histological and immunohistochemical features. MSC/chondrocyte co-aggregation in comparison to MSC or chondrocyte aggregation could increase EV yield during dynamic 3D culture by spinner flasks. Although MSC-ag-, Cho-ag- and Co-ag-derived EVs could induce chondrogenesis similar to transforming growth factor-beta during in vitro study, Co-ag-EV could more effectively prevent OA progression than MSC-ag- and Cho-ag-EVs. Our study demonstrated that EVs harvested from the co-aggregation of MSCs and chondrocytes could be considered as a new therapeutic potential for OA treatment.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Kamali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Hosseinzadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
128
|
Wang Y, Fan A, Lu L, Pan Z, Ma M, Luo S, Liu Z, Yang L, Cai J, Yin F. Exosome modification to better alleviates endoplasmic reticulum stress induced chondrocyte apoptosis and osteoarthritis. Biochem Pharmacol 2022; 206:115343. [DOI: 10.1016/j.bcp.2022.115343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022]
|
129
|
MSC-EV therapy for bone/cartilage diseases. Bone Rep 2022; 17:101636. [DOI: 10.1016/j.bonr.2022.101636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [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
|
130
|
Liu C, Li Y, Han G. Advances of Mesenchymal Stem Cells Released Extracellular Vesicles in Periodontal Bone Remodeling. DNA Cell Biol 2022; 41:935-950. [PMID: 36315196 DOI: 10.1089/dna.2022.0359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles that include exosomes, microvesicles, and apoptotic bodies; they interact with target cell surface receptors and transport contents, including mRNA, proteins, and enzymes into the cytoplasm of target cells to function. The biological fingerprints of EVs practically mirror those of the parental cells they originated from. In the bone remodeling microenvironment, EVs could act on osteoblasts to regulate the bone formation, promote osteoclast differentiation, and regulate bone resorption. Therefore, there have been many attempts wherein EVs were used to achieve targeted therapy in bone-related diseases. Periodontitis, a common bacterial infectious disease, could cause severe alveolar bone resorption, resulting in tooth loss, whereas research on periodontal bone regeneration is also an urgent question. Therefore, EVs-related studies are important for periodontal bone remodeling. In this review, we summarize the current knowledge of mesenchymal stem cell-EVs involved in periodontal bone remodeling and explore the functional gene expression through a comparative analysis of transcriptomic content.
Collapse
Affiliation(s)
- Chaoran Liu
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Yanan Li
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Guanghong Han
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
131
|
Kuppa SS, Kim HK, Kang JY, Lee SC, Seon JK. Role of Mesenchymal Stem Cells and Their Paracrine Mediators in Macrophage Polarization: An Approach to Reduce Inflammation in Osteoarthritis. Int J Mol Sci 2022; 23:13016. [PMID: 36361805 PMCID: PMC9658630 DOI: 10.3390/ijms232113016] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoarthritis (OA) is a low-grade inflammatory disorder of the joints that causes deterioration of the cartilage, bone remodeling, formation of osteophytes, meniscal damage, and synovial inflammation (synovitis). The synovium is the primary site of inflammation in OA and is frequently characterized by hyperplasia of the synovial lining and infiltration of inflammatory cells, primarily macrophages. Macrophages play a crucial role in the early inflammatory response through the production of several inflammatory cytokines, chemokines, growth factors, and proteinases. These pro-inflammatory mediators are activators of numerous signaling pathways that trigger other cytokines to further recruit more macrophages to the joint, ultimately leading to pain and disease progression. Very few therapeutic alternatives are available for treating inflammation in OA due to the condition's low self-healing capacity and the lack of clear diagnostic biomarkers. In this review, we opted to explore the immunomodulatory properties of mesenchymal stem cells (MSCs) and their paracrine mediators-dependent as a therapeutic intervention for OA, with a primary focus on the practicality of polarizing macrophages as suppression of M1 macrophages and enhancement of M2 macrophages can significantly reduce OA symptoms.
Collapse
Affiliation(s)
- Sree Samanvitha Kuppa
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Hyung Keun Kim
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Ju Yeon Kang
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Seok Cheol Lee
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| | - Jong Keun Seon
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501-757, Korea
| |
Collapse
|
132
|
McLaughlin C, Datta P, Singh YP, Lo A, Horchler S, Elcheva IA, Ozbolat IT, Ravnic DJ, Koduru SV. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Therapeutic Use and in Bioengineering Applications. Cells 2022; 11:3366. [PMID: 36359762 PMCID: PMC9657427 DOI: 10.3390/cells11213366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 07/25/2023] Open
Abstract
Extracellular vesicles (EVs) are small lipid bilayer-delimited particles that are naturally released from cells into body fluids, and therefore can travel and convey regulatory functions in the distal parts of the body. EVs can transmit paracrine signaling by carrying over cytokines, chemokines, growth factors, interleukins (ILs), transcription factors, and nucleic acids such as DNA, mRNAs, microRNAs, piRNAs, lncRNAs, sn/snoRNAs, mtRNAs and circRNAs; these EVs travel to predecided destinations to perform their functions. While mesenchymal stem cells (MSCs) have been shown to improve healing and facilitate treatments of various diseases, the allogenic use of these cells is often accompanied by serious adverse effects after transplantation. MSC-produced EVs are less immunogenic and can serve as an alternative to cellular therapies by transmitting signaling or delivering biomaterials to diseased areas of the body. This review article is focused on understanding the properties of EVs derived from different types of MSCs and MSC-EV-based therapeutic options. The potential of modern technologies such as 3D bioprinting to advance EV-based therapies is also discussed.
Collapse
Affiliation(s)
- Caroline McLaughlin
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, West Bengal 700054, India
| | - Yogendra P. Singh
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Alexis Lo
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Summer Horchler
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Irina A. Elcheva
- Department of Pediatrics, Hematology/Oncology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Ibrahim T. Ozbolat
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Dino J. Ravnic
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Srinivas V. Koduru
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
133
|
Breakthrough of extracellular vesicles in pathogenesis, diagnosis and treatment of osteoarthritis. Bioact Mater 2022; 22:423-452. [PMID: 36311050 PMCID: PMC9588998 DOI: 10.1016/j.bioactmat.2022.10.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent whole-joint disease that causes disability and pain and affects a patient's quality of life. However, currently, there is a lack of effective early diagnosis and treatment. Although stem cells can promote cartilage repair and treat OA, problems such as immune rejection and tumorigenicity persist. Extracellular vesicles (EVs) can transmit genetic information from donor cells and mediate intercellular communication, which is considered a functional paracrine factor of stem cells. Increasing evidences suggest that EVs may play an essential and complex role in the pathogenesis, diagnosis, and treatment of OA. Here, we introduced the role of EVs in OA progression by influencing inflammation, metabolism, and aging. Next, we discussed EVs from the blood, synovial fluid, and joint-related cells for diagnosis. Moreover, we outlined the potential of modified and unmodified EVs and their combination with biomaterials for OA therapy. Finally, we discuss the deficiencies and put forward the prospects and challenges related to the application of EVs in the field of OA.
Collapse
|
134
|
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
|
135
|
AbuBakr N, Fares AE, Mostafa A, Farag DB. Mesenchymal stem cells-derived microvesicles versus platelet-rich plasma in the treatment of monoiodoacetate-induced temporomandibular joint osteoarthritis in Albino rats. Heliyon 2022; 8:e10857. [PMID: 36212013 PMCID: PMC9539788 DOI: 10.1016/j.heliyon.2022.e10857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/14/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022] Open
Abstract
Temporomandibular joint osteoarthritis (TMJ-OA) is a serious disease, designated by severe joint pain and dysfunction. Limitations of current therapeutics have led to an increased interest in regenerative strategies. Recently, the non-surgical treatment of OA has seen increased use of biologic injectable therapies like mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP). Although these biotherapies represent an admirable effort, more studies are necessary to determine their efficacy. Thus, the aim of this study was to assess the curative potential of a single intra-articular injection of bone marrow MSCs-derived microvesicles (BM-MSCs-MVs) versus a single intra-articular injection of PRP in monoiodoacetate (MIA)-induced TMJ-OA model in Albino rats. Forty-eight male rats were used. A single intra-articular unilateral MIA injection was utilized to induce TMJ-OA. One week post induction, rats were sorted into 3 groups (16 rats each): group (I): received no treatment, groups (II) & (III): received BM-MSCs-MVs and PRP respectively. Scarification was done at 2 and 4 weeks from onset of treatment. Histological changes of the condylar TMJ were examined with H&E staining. Expression of IL-1β, TNF-α, NF-κB, MMP-13, MMP-3, and collagen ΙΙ markers was detected using real-time PCR. Histologically, the osteoarthritic group exhibited degenerated condylar tissues which were aggravated at 4 weeks. Oppositely, a marked improvement in the condylar TMJ histology was noticed in both the BM-MSCs-MVs-and PRP-treated groups at both time intervals. Additionally, the treated groups showed a decrease in IL-1β, TNF-α, NF-κB, MMP-13 and MMP-3 and an increase in collagen ΙΙ genes expression in contrast to the untreated group. Moreover, this difference was significant in the BM-MSCs-MVs group as compared to the PRP-treated group. Our results concluded that BM-MSCs-MVs as well as PRP treatments were able to target the key pathological features in OA, mainly inflammation and matrix degradation, and helped in restoring condylar structure in TMJ-OA rat model. However, BM-MSCs-MVs treatment exhibited more efficient therapeutic potential as compared to PRP treatment.
Collapse
|
136
|
Zhuang Y, Jiang S, Yuan C, Lin K. The potential therapeutic role of extracellular vesicles in osteoarthritis. Front Bioeng Biotechnol 2022; 10:1022368. [PMID: 36185451 PMCID: PMC9523151 DOI: 10.3389/fbioe.2022.1022368] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a worldwide and disabling disease, which cause severe pain and heavy socioeconomic burden. However, pharmacologic or surgical therapies cannot mitigate OA progression. Mesenchymal stem cells (MSCs) therapy has emerged as potential approach for OA treatment, while the immunogenicity and ethical audit of cell therapy are unavoidable. Compared with stem cell strategy, EVs induce less immunological rejection, and they are more stable for storage and in vivo application. MSC-EVs-based therapy possesses great potential in regulating inflammation and promoting cartilage matrix reconstruction in OA treatment. To enhance the therapeutic effect, delivery efficiency, tissue specificity and safety, EVs can be engineered via different modification strategies. Here, the application of MSC-EVs in OA treatment and the potential underlying mechanism were summarized. Moreover, EV modification strategies including indirect MSC modification and direct EV modification were reviewed.
Collapse
Affiliation(s)
- Yu Zhuang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shengjie Jiang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, Shanghai, China
- Department of Dental Implant, The Affiliated Stomatological Hospital of Xuzhou Medical University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Changyong Yuan, ; Kaili Lin,
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: Changyong Yuan, ; Kaili Lin,
| |
Collapse
|
137
|
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
|
138
|
Turner ME, Bartoli‐Leonard F, Aikawa E. Small particles with large impact: Insights into the unresolved roles of innate immunity in extracellular vesicle‐mediated cardiovascular calcification. Immunol Rev 2022; 312:20-37. [DOI: 10.1111/imr.13134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mandy E Turner
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Francesca Bartoli‐Leonard
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
- Division of Cardiovascular Medicine Department of Medicine Center for Excellence in Vascular Biology Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
139
|
The Effect of Human Bone Marrow Mesenchymal Stem Cell-Derived Exosomes on Cartilage Repair in Rabbits. Stem Cells Int 2022; 2022:5760107. [PMID: 36117721 PMCID: PMC9477595 DOI: 10.1155/2022/5760107] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/22/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have shown chondroprotective effects in cartilage repair. However, side effects caused by MSC treatment limit their application in clinic. As a cell-free therapy, MSC-derived exosomes (EXOs) have attracted much more attention in recent years. In the present study, we prepared EXOs from human bone marrow mesenchymal stem cells (hBMSCs) and examined their therapeutic potentials in cartilage repair. Our results showed that the prepared extracellular vesicles exhibit classical features of EXOs, such as cup-like shape, around 100 nm diameter, positive protein markers (CD81, TSG101, and Flotillin 1), and ability of internalization. In primary chondrocytes, the treatment of hBMSC-EXOs markedly increases cell viability and proliferation in a dose-dependent manner. Moreover, wound healing assay showed that hBMSC-EXOs accelerate cell migration in primary chondrocytes. JC-1 staining revealed that the mitochondrial membrane potential was enhanced by hBMSC-EXOs, indicating cell apoptosis was decreased in the presence of hBMSC-EXOs. In rabbits with articular cartilage defects, local administration with hBMSC-EXOs facilitates cartilage regeneration as evidenced by gross view and hematoxylin-eosin (H&E) and Saf-O/Fast Green staining. In addition, the International Cartilage Repair Society (ICRS) score was increased by the application of hBMSC-EXOs. Overall, our data indicate that the treatment with hBMSC-EXOs is a suitable cell-free therapy for treating cartilage defects, and these benefits are likely due to improved cell proliferation and migration in chondrocytes.
Collapse
|
140
|
Lu Y, Yang Y, Liu S, Ge S. Biomaterials constructed for MSC-derived extracellular vesicle loading and delivery—a promising method for tissue regeneration. Front Cell Dev Biol 2022; 10:898394. [PMID: 36092710 PMCID: PMC9454000 DOI: 10.3389/fcell.2022.898394] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have become the preferred seed cells for tissue regeneration. Nevertheless, due to their immunogenicity and tumorigenicity, MSC transplantation remains questionable. Extracellular vesicles (EVs) derived from MSCs are becoming a promising substitute for MSCs. As a route of the MSC paracrine, EVs have a nano-sized and bilayer lipid-enclosed structure, which can guarantee the integrity of their cargoes, but EVs cannot obtain full function in vivo because of the rapid biodegradation and clearance by phagocytosis. To improve the efficacy and targeting of EVs, methods have been proposed and put into practice, especially engineered vesicles and EV-controlled release systems. In particular, EVs can be cell or tissue targeting because they have cell-specific ligands on their surfaces, but their targeting ability may be eliminated by the biodegradation of the phagocytic system during circulation. Novel application strategies have been proposed beyond direct injecting. EV carriers such as biodegradable hydrogels and other loading systems have been applied in tissue regeneration, and EV engineering is also a brand-new method for higher efficacy. In this review, we distinctively summarize EV engineering and loading system construction methods, emphasizing targeting modification methods and controlled release systems for EVs, which few literature reviews have involved.
Collapse
Affiliation(s)
- Yu Lu
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu Yang
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| | - Shaohua Ge
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Shaohua Ge,
| |
Collapse
|
141
|
Hao D, Lu L, Song H, Duan Y, Chen J, Carney R, Li JJ, Zhou P, Nolta J, Lam KS, Leach JK, Farmer DL, Panitch A, Wang A. Engineered extracellular vesicles with high collagen-binding affinity present superior in situ retention and therapeutic efficacy in tissue repair. Theranostics 2022; 12:6021-6037. [PMID: 35966577 PMCID: PMC9373818 DOI: 10.7150/thno.70448] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 07/24/2022] [Indexed: 01/26/2023] Open
Abstract
Although stem cell-derived extracellular vesicles (EVs) have remarkable therapeutic potential for various diseases, the therapeutic efficacy of EVs is limited due to their degradation and rapid diffusion after administration, hindering their translational applications. Here, we developed a new generation of collagen-binding EVs, by chemically conjugating a collagen-binding peptide SILY to EVs (SILY-EVs), which were designed to bind to collagen in the extracellular matrix (ECM) and form an EV-ECM complex to improve EVs' in situ retention and therapeutic efficacy after transplantation. Methods: SILY was conjugated to the surface of mesenchymal stem/stromal cell (MSC)-derived EVs by using click chemistry to construct SILY-EVs. Nanoparticle tracking analysis (NTA), ExoView analysis, cryogenic electron microscopy (cryo-EM) and western-blot analysis were used to characterize the SILY-EVs. Fluorescence imaging (FLI), MTS assay, ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to evaluate the collagen binding and biological functions of SILY-EVs in vitro. In a mouse hind limb ischemia model, the in vivo imaging system (IVIS), laser doppler perfusion imaging (LDPI), micro-CT, FLI and RT-qPCR were used to determine the SILY-EV retention, inflammatory response, blood perfusion, gene expression, and tissue regeneration. Results:In vitro, the SILY conjugation significantly enhanced EV adhesion to the collagen surface and did not alter the EVs' biological functions. In the mouse hind limb ischemia model, SILY-EVs presented longer in situ retention, suppressed inflammatory responses, and significantly augmented muscle regeneration and vascularization, compared to the unmodified EVs. Conclusion: With the broad distribution of collagen in various tissues and organs, SILY-EVs hold promise to improve the therapeutic efficacy of EV-mediated treatment in a wide range of diseases and disorders. Moreover, SILY-EVs possess the potential to functionalize collagen-based biomaterials and deliver therapeutic agents for regenerative medicine applications.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Lu Lu
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Hengyue Song
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Yixin Duan
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Jianing Chen
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Randy Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Ping Zhou
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Jan Nolta
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - J. Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Diana L Farmer
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Alyssa Panitch
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Aijun Wang
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
142
|
Lv B, Cheng Z, Yu Y, Chen Y, Gan W, Li S, Zhao K, Yang C, Zhang Y. Therapeutic perspectives of exosomes in glucocorticoid-induced osteoarthrosis. Front Surg 2022; 9:836367. [PMID: 36034358 PMCID: PMC9405187 DOI: 10.3389/fsurg.2022.836367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022] Open
Abstract
Exosomes are widely involved in a variety of physiological and pathological processes. These important roles are also hidden in the physiological processes related to bone. Chondrocytes, osteoblasts, synovial fibroblasts, and bone marrow mesenchymal stem cells produce and secrete exosomes, thereby affecting the biology process of target cells. Furthermore, in the primary pathogenesis of osteoarthrosis induced by steroid hormones, mainly involve glucocorticoid (GC), the exosomes have also widely participated. Therefore, exosomes may also play an important role in glucocorticoid-induced osteoarthrosis and serve as a promising treatment for early intervention of osteoarthrosis in addition to playing a regulatory role in malignant tumors. This review summarizes the previous results on this direction, systematically combs the role and therapeutic potential of exosomes in GC-induced osteoarthrosis, discusses the potential role of exosomes in the treatment and prevention of GC-induced osteoarthrosis, and reveals the current challenges we confronted.
Collapse
Affiliation(s)
- Bin Lv
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| | | | | | | | | | | | - Kangcheng Zhao
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| | - Cao Yang
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| | - Yukun Zhang
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| |
Collapse
|
143
|
Fan WJ, Liu D, Pan LY, Wang WY, Ding YL, Zhang YY, Ye RX, Zhou Y, An SB, Xiao WF. Exosomes in osteoarthritis: Updated insights on pathogenesis, diagnosis, and treatment. Front Cell Dev Biol 2022; 10:949690. [PMID: 35959489 PMCID: PMC9362859 DOI: 10.3389/fcell.2022.949690] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 07/04/2022] [Indexed: 01/09/2023] Open
Abstract
Osteoarthritis (OA) has remained a prevalent public health problem worldwide over the past decades. OA is a global challenge because its specific pathogenesis is unclear, and no effective disease-modifying drugs are currently available. Exosomes are small and single-membrane vesicles secreted via the formation of endocytic vesicles and multivesicular bodies (MVBs), which are eventually released when MVBs fuse with the plasma membrane. Exosomes contain various integral surface proteins derived from cells, intercellular proteins, DNAs, RNAs, amino acids, and metabolites. By transferring complex constituents and promoting macrophages to generate chemokines and proinflammatory cytokines, exosomes function in pathophysiological processes in OA, including local inflammation, cartilage calcification and degradation of osteoarthritic joints. Exosomes are also detected in synovial fluid and plasma, and their levels continuously change with OA progression. Thus, exosomes, specifically exosomal miRNAs and lncRNAs, potentially represent multicomponent diagnostic biomarkers for OA. Exosomes derived from various types of mesenchymal stem cells and other cell or tissue types affect angiogenesis, inflammation, and bone remodeling. These exosomes exhibit promising capabilities to restore OA cartilage, attenuate inflammation, and balance cartilage matrix formation and degradation, thus demonstrating therapeutic potential in OA. In combination with biocompatible and highly adhesive materials, such as hydrogels and cryogels, exosomes may facilitate cartilage tissue engineering therapies for OA. Based on numerous recent studies, we summarized the latent mechanisms and clinical value of exosomes in OA in this review.
Collapse
Affiliation(s)
- Wen-Jin Fan
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Lin-Yuan Pan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Wei-Yang Wang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yi-Lan Ding
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yue-Yao Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Rui-Xi Ye
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yang Zhou
- Department of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Yang Zhou, ; Sen-Bo An, ; Wen-Feng Xiao,
| | - Sen-Bo An
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China,*Correspondence: Yang Zhou, ; Sen-Bo An, ; Wen-Feng Xiao,
| | - Wen-Feng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Yang Zhou, ; Sen-Bo An, ; Wen-Feng Xiao,
| |
Collapse
|
144
|
BMSC-derived exosomes promote tendon-bone healing after anterior cruciate ligament reconstruction by regulating M1/M2 macrophage polarization in rats. Stem Cell Res Ther 2022; 13:295. [PMID: 35841008 PMCID: PMC9284827 DOI: 10.1186/s13287-022-02975-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/06/2022] [Indexed: 11/22/2022] Open
Abstract
Background Recent studies have shown that bone marrow stromal cell-derived exosomes (BMSC-Exos) can be used for tissue repair. However, whether the BMSC-Exos can promote tendon-bone healing after anterior cruciate ligament reconstruction (ACLR) is still unclear. In this study, we observed in vivo and in vitro the effect of rat BMSC-Exos on tendon-bone healing after ACLR and its possible mechanism. Methods Highly expressed miRNAs in rat BMSC-Exos were selected by bioinformatics and verified in vitro. The effect of overexpressed miRNA in BMSC-Exos on M2 macrophage polarization was observed. A rat model of ACLR was established. The experimental components were divided into three groups: the control group, the BMSC-Exos group, and the BMSC-Exos with miR-23a-3p overexpression (BMSC-Exos mimic) group. Biomechanical tests, micro-CT, and histological staining were performed for analysis. Results Bioinformatics analysis showed that miR-23a-3p was highly expressed in rat BMSC-Exos and could target interferon regulatory factor 1 (IRF1, a crucial regulator in M1 macrophage polarization). In vitro, compared with the control group or the BMSC-Exos group, the BMSC-Exos mimic more significantly promoted the polarization of macrophages from M1 to M2. In vivo, at 2 weeks, the number of M2 macrophages in the early local stage of ACLR was significantly increased in the BMSC-Exos mimic group; at 4 and 8 weeks, compared with the control group or the BMSC-Exos group, the bone tunnels of the tibia and femur sides of the rats in the BMSC-Exos mimic group were significantly smaller, the interface between the graft and the bone was narrowed, the bone volume/total volume ratio (BV/TV) increased, the collagen type II alpha 1 level increased, and the mechanical strength increased. Conclusions BMSC-Exos promoted M1 macrophage to M2 macrophage polarization via miR-23a-3p, reduced the early inflammatory reaction at the tendon-bone interface, and promoted early healing after ACLR. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02975-0.
Collapse
|
145
|
Yu H, Huang Y, Yang L. Research progress in the use of mesenchymal stem cells and their derived exosomes in the treatment of osteoarthritis. Ageing Res Rev 2022; 80:101684. [PMID: 35809775 DOI: 10.1016/j.arr.2022.101684] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/08/2022] [Accepted: 07/05/2022] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA), as a common orthopedic disease with cartilage injury as its main pathological feature, has a complex pathogenesis and existing medical technology remains unable to reverse the progress of cartilage degeneration caused thereby. In recent years, mesenchymal stem cells (MSCs) and their secreted exosomes have become a focus of research into cartilage regeneration. MSCs have the potential to differentiate into a variety of cells. Under specific conditions, they can be promoted to differentiate into chondrocytes and maintain the function and stability of chondrocytes. Exosomes secreted by MSCs, as an intercellular messenger, can treat OA in a variety of ways through bioactive factors carried therewith, such as protein, lipid, mRNA, and miRNA. This study reviewed the application of MSCs and their exosomes from different sources in the prevention of OA, which provides a new idea for the treatment of OA.
Collapse
Affiliation(s)
- Hongxia Yu
- Departments of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Yuling Huang
- Departments of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Lina Yang
- Departments of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
146
|
Iravani K, Mehrabani D, Doostkam A, Azarpira N, Iranpour P, Bahador M, Mehravar S. Use of MRI to Assess the Regenerative Effects of Adipose Tissue Derived Mesenchymal Stem Cells in a Rabbit Cartilaginous Laryngeal Defect Model. Curr Ther Res Clin Exp 2022; 97:100682. [PMID: 35959231 PMCID: PMC9361331 DOI: 10.1016/j.curtheres.2022.100682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
Background Stenosis and scar formation after repair of laryngeal tissue defects are serious problems that can significantly influence a patient's quality of life. Objective In this study, we evaluated the use of magnetic resonance imaging to assess the efficacy of adipose tissue-derived mesenchymal stem cells (ASCs) on cartilaginous regeneration in an experimental rabbit model. Methods Ten male white Dutch rabbits each had a 5 mm cartilaginous defect created surgically in the right and left thyroid lamina. On the right side, ASCs labeled with iron oxide particles were infused. As a control, the left side was left untreated. Repair of the defects were then evaluated by direct observation, histological evaluation, and magnetic resonance imaging monitoring done on days 1, 7, 14, and 28. Results Histological examination revealed that compared with control, transplanted ASCs significantly increased cartilage regeneration (P ˂ 0.001), reduced inflammation (P ˂ 0.001), and fibrosis (P = 0.050). Magnetic resonance imaging tracking showed accurate placement and viability of the infused ASCs, as evidenced by low signal intensity onT2 weighted images at the level of the right thyroid cartilage. Conclusions Infusion of ASCs improved laryngeal regeneration of surgically induced cartilaginous defects while decreasing fibrous tissue formation in this in vivo rabbit model. Furthermore, magnetic resonance imaging was shown to be a useful, noninvasive method to track correct ASCs placement and viability in cartilage regeneration in this animal model.
Collapse
Affiliation(s)
- Kamyar Iravani
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
- Address correspondence to: Kamyar Iravani, MD, Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Khalili Hospital, Khalili St, Shiraz, Iran, Postal code: 71936-16641.
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Doostkam
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz Institute of Stem Cell and Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooya Iranpour
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Bahador
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Mehravar
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
147
|
Loh JT, Zhang B, Teo JKH, Lai RC, Choo ABH, Lam KP, Lim SK. Mechanism for the attenuation of neutrophil and complement hyperactivity by MSC exosomes. Cytotherapy 2022; 24:711-719. [PMID: 35177337 PMCID: PMC8843421 DOI: 10.1016/j.jcyt.2021.12.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/08/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022]
Abstract
Complements and neutrophils are two key players of the innate immune system that are widely implicated as drivers of severe COVID-19 pathogenesis, as evident by the direct correlation of respiratory failure and mortality with elevated levels of terminal complement complex C5b-9 and neutrophils. In this study, we identified a feed-forward loop between complements and neutrophils that could amplify and perpetuate the cytokine storm seen in severe SARS-CoV-2-infected patients. We observed for the first time that the terminal complement activation complex C5b-9 directly triggered neutrophil extracellular trap (NET) release and interleukin (IL)-17 production by neutrophils. This is also the first report that the production of NETs and IL-17 induced by C5b-9 assembly on neutrophils could be abrogated by mesenchymal stem cell (MSC) exosomes. Neutralizing anti-CD59 antibodies abolished this abrogation. Based on our findings, we hypothesize that MSC exosomes could alleviate the immune dysregulation in acute respiratory failure, such as that observed in severe COVID-19 patients, by inhibiting complement activation through exosomal CD59, thereby disrupting the feed-forward loop between complements and neutrophils to inhibit the amplification and perpetuation of inflammation during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jia Tong Loh
- Singapore Immunology Network, Agency for Science, Technology and Research, Republic of Singapore
| | - Bin Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Republic of Singapore
| | - Joey Kay Hui Teo
- Singapore Immunology Network, Agency for Science, Technology and Research, Republic of Singapore
| | - Ruenn Chai Lai
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Republic of Singapore
| | - Andre Boon Hwa Choo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Republic of Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology and Research, Republic of Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore; School of Biological Sciences, College of Science, Nanyang Technological University, Republic of Singapore.
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Republic of Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore.
| |
Collapse
|
148
|
Bhujel B, Shin HE, Choi DJ, Han I. Mesenchymal Stem Cell-Derived Exosomes and Intervertebral Disc Regeneration: Review. Int J Mol Sci 2022; 23:7306. [PMID: 35806304 PMCID: PMC9267028 DOI: 10.3390/ijms23137306] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 02/07/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) is a common cause of lower back pain (LBP), which burdens individuals and society as a whole. IVDD occurs as a result of aging, mechanical trauma, lifestyle factors, and certain genetic abnormalities, leads to loss of nucleus pulposus, alteration in the composition of the extracellular matrix, excessive oxidative stress, and inflammation in the intervertebral disc. Pharmacological and surgical interventions are considered a boon for the treatment of IVDD, but the effectiveness of those strategies is limited. Mesenchymal stem cells (MSCs) have recently emerged as a possible promising regenerative therapy for IVDD due to their paracrine effect, restoration of the degenerated cells, and capacity for differentiation into disc cells. Recent investigations have shown that the pleiotropic effect of MSCs is not related to differentiation capacity but is mediated by the secretion of soluble paracrine factors. Early studies have demonstrated that MSC-derived exosomes have therapeutic potential for treating IVDD by promoting cell proliferation, tissue regeneration, modulation of the inflammatory response, and reduced apoptosis. This paper highlights the current state of MSC-derived exosomes in the field of treatment of IVDD with further possible future developments, applications, and challenges.
Collapse
Affiliation(s)
- Basanta Bhujel
- Department of Biomedical Science, College of Life Sciences, CHA University, Seongnam-si 13496, Korea; (B.B.); (H.-E.S.)
| | - Hae-Eun Shin
- Department of Biomedical Science, College of Life Sciences, CHA University, Seongnam-si 13496, Korea; (B.B.); (H.-E.S.)
| | - Dong-Jun Choi
- Department of Medicine, CHA Univerity School of Medicine, Seongnam-si 13496, Korea;
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Korea
| |
Collapse
|
149
|
Karimi B, Dehghani Firoozabadi A, Peymani M, Ghaedi K. Circulating long noncoding RNAs as novel bio-tools: Focus on autoimmune diseases. Hum Immunol 2022; 83:618-627. [PMID: 35717260 DOI: 10.1016/j.humimm.2022.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/04/2022]
Abstract
Long non-coding RNAs (lncRNAs) are an emerging class of non-coding RNAs that do not encode proteins. These RNAs have various essential regulatory functions. Irregular expression of lncRNAs has been related to the pathological process of varied diseases, and are considered promising diagnostic biomarkers. LncRNAs can release into the circulation and be stable in body fluids as circulating lncRNAs. A subset of circulating lncRNAs that exist in exosomes are referred to as exosomal lncRNA molecules. These lncRNAs are highly stable and resist RNases. Exosomes have captured a great deal of attention due to their involvement in regulating communications between cells. In conditions of autoimmune disease, exosomes play critical roles in the pathological processes. In this context, circulating lncRNAs have been shown to modulate the immune response and indicated as prognosis and diagnostic biomarkers for autoimmune diseases. This review highlights the role of circulating lncRNAs (particularly exosomal) as diagnostic biomarkers for autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, psoriasis, and Sjögren's syndrome.
Collapse
Affiliation(s)
- Bahareh Karimi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | | | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Kamran Ghaedi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
150
|
Wong KL, Zhang S, Tan SSH, Cheow YA, Lai RC, Lim SK, Hui JHP, Toh WS. Mesenchymal Stem Cell Exosomes Promote Growth Plate Repair and Reduce Limb-Length Discrepancy in Young Rats. J Bone Joint Surg Am 2022; 104:1098-1106. [PMID: 35175995 DOI: 10.2106/jbjs.21.00789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The objective of this study was to examine the therapeutic effects of human mesenchymal stromal/stem cell (MSC) exosomes in a rat model of growth plate injury. METHODS A growth plate defect was surgically created on the distal part of the right femur of 40 female Sprague-Dawley rats. A single intra-articular injection of 100 µg of MSC exosomes in 100 µL of phosphate-buffered saline solution (PBS), or an equivalent volume of PBS alone, was administered to the right knee immediately after surgery. At 4 and 8 weeks post-treatment, limb length was measured with micro-CT, and tissue repair was assessed with histological, immunohistochemical, and histomorphometric analyses. RESULTS A single injection of MSC exosomes significantly increased limb length from 3.29 ± 0.07 cm at 4 weeks to 3.37 ± 0.11 cm at 8 weeks (p = 0.047). However, no improvement in limb length was observed in the PBS control group. The limb-length discrepancy between the involved limb and the contralateral limb in the exosome-treated group was significantly less than the discrepancy in the PBS-treated group at both 4 weeks (2.52% ± 1.30% versus 4.11% ± 0.93%; p = 0.006) and 8 weeks (5.27% ± 2.11% versus 8.06% ± 2.56%; p = 0.016). Consistent with the reduced limb-length discrepancy, the exosome-treated defects displayed significantly more chondrocytes (p < 0.05) and a higher area percentage with deposition of sulphated glycosaminoglycan (p < 0.05) and collagen II (p < 0.05) than PBS-treated defects at 8 weeks. However, bone bridge formation was not inhibited in either group. CONCLUSIONS A single intra-articular injection of MSC exosomes significantly enhanced physeal repair and reduced limb-length discrepancy but did not inhibit bone-bridge formation. CLINICAL RELEVANCE This proof-of-concept study demonstrates for the first time the potential use of MSC exosomes as a minimally invasive cell-free therapeutic to promote physeal repair and reduce limb-length discrepancy following growth plate injuries.
Collapse
Affiliation(s)
- Keng Lin Wong
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Sengkang General Hospital, Singhealth, Singapore
| | - Shipin Zhang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Faculty of Dentistry, National University of Singapore, Singapore
| | - Sharon Si Heng Tan
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yi Ann Cheow
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Ruenn Chai Lai
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - Wei Seong Toh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Faculty of Dentistry, National University of Singapore, Singapore.,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Integrative Sciences and Engineering Program, NUS Graduate School, National University of Singapore, Singapore
| |
Collapse
|