201
|
Skeletal Muscle Regeneration by the Exosomes of Adipose Tissue-Derived Mesenchymal Stem Cells. Curr Issues Mol Biol 2021; 43:1473-1488. [PMID: 34698065 PMCID: PMC8929094 DOI: 10.3390/cimb43030104] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/17/2022] Open
Abstract
Profound skeletal muscle loss can lead to severe disability and cosmetic deformities. Mesenchymal stem cell (MSC)-derived exosomes have shown potential as an effective therapeutic tool for tissue regeneration. This study aimed to determine the regenerative capacity of MSC-derived exosomes for skeletal muscle regeneration. Exosomes were isolated from human adipose tissue-derived MSCs (AD-MSCs). The effects of MSC-derived exosomes on satellite cells were investigated using cell viability, relevant genes, and protein analyses. Moreover, NOD-SCID mice were used and randomly assigned to the healthy control (n = 4), muscle defect (n = 6), and muscle defect + exosome (n = 6) groups. Muscle defects were created using a biopsy punch on the quadriceps of the hind limb. Four weeks after the surgery, the quadriceps muscles were harvested, weighed, and histologically analyzed. MSC-derived exosome treatment increased the proliferation and expression of myocyte-related genes, and immunofluorescence analysis for myogenin revealed a similar trend. Histologically, MSC-derived exosome-treated mice showed relatively preserved shapes and sizes of the muscle bundles. Immunohistochemical staining revealed greater expression of myogenin and myoblast determination protein 1 in the MSC-derived exosome-treated group. These results indicate that exosomes extracted from AD-MSCs have the therapeutic potential for skeletal muscle regeneration.
Collapse
|
202
|
Ng CY, Chai JY, Foo JB, Mohamad Yahaya NH, Yang Y, Ng MH, Law JX. Potential of Exosomes as Cell-Free Therapy in Articular Cartilage Regeneration: A Review. Int J Nanomedicine 2021; 16:6749-6781. [PMID: 34621125 PMCID: PMC8491788 DOI: 10.2147/ijn.s327059] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/22/2021] [Indexed: 12/20/2022] Open
Abstract
Treatment of cartilage defects such as osteoarthritis (OA) and osteochondral defect (OCD) remains a huge clinical challenge in orthopedics. OA is one of the most common chronic health conditions and is mainly characterized by the degeneration of articular cartilage, shown in the limited capacity for intrinsic repair. OCD refers to the focal defects affecting cartilage and the underlying bone. The current OA and OCD management modalities focus on symptom control and on improving joint functionality and the patient’s quality of life. Cell-based therapy has been evaluated for managing OA and OCD, and its chondroprotective efficacy is recognized mainly through paracrine action. Hence, there is growing interest in exploiting extracellular vesicles to induce cartilage regeneration. In this review, we explore the in vivo evidence of exosomes on cartilage regeneration. A total of 29 in vivo studies from the PubMed and Scopus databases were identified and analyzed. The studies reported promising results in terms of in vivo exosome delivery and uptake; improved cartilage morphological, histological, and biochemical outcomes; enhanced subchondral bone regeneration; and improved pain behavior following exosome treatment. In addition, exosome therapy is safe, as the included studies documented no significant complications. Modifying exosomal cargos further increased the cartilage and subchondral bone regeneration capacity of exosomes. We conclude that exosome administration is a potent cell-free therapy for alleviating OA and OCD. However, additional studies are needed to confirm the therapeutic potential of exosomes and to identify the standard protocol for exosome-based therapy in OA and OCD management.
Collapse
Affiliation(s)
- Chiew Yong Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| | - Jia Ying Chai
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Selangor, Malaysia.,Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, 47500, Malaysia
| | - Nor Hamdan Mohamad Yahaya
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, 56000, Malaysia
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| |
Collapse
|
203
|
Wu J, Piao Y, Liu Q, Yang X. Platelet-rich plasma-derived extracellular vesicles: A superior alternative in regenerative medicine? Cell Prolif 2021; 54:e13123. [PMID: 34609779 PMCID: PMC8666280 DOI: 10.1111/cpr.13123] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/13/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023] Open
Abstract
Platelet-rich plasma (PRP), due to its promising therapeutic properties, has been used in regenerative medicine for more than 30 years and numerous encouraging outcomes have been obtained. Currently, by benefiting from new insights into PRP mechanisms and the excellent performance of extracellular vesicles (EVs) in the field of tissue repair and regeneration, studies have found that a large number of EVs released from activated platelets also participate in the regulation of tissue repair. A growing number of preclinical studies are exploring the functions of PRP-derived EVs (PRP-EVs), especially in tissue regeneration. Here, we summarize the latest progress in PRP-EVs as a superior alternative cell-free therapeutic strategy in regenerative medicine, clarify their underlying molecular mechanisms, and discuss the advantages and limitations of the upcoming clinical applications. This review highlights the potential of PRP-EVs to replace the application of PRP or even become a superior alternative in regenerative medicine.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopaedics, The Second Hospital, Jilin University, Changchun, China
| | - Yingxin Piao
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Qinyi Liu
- Department of Orthopaedics, The Second Hospital, Jilin University, Changchun, China
| | - Xiaoyu Yang
- Department of Orthopaedics, The Second Hospital, Jilin University, Changchun, China
| |
Collapse
|
204
|
Ghasemian SO. Application of Exosomes-Derived Mesenchymal Stem Cells in Treatment of Fungal Diseases: From Basic to Clinical Sciences. FRONTIERS IN FUNGAL BIOLOGY 2021; 2:736093. [PMID: 37744094 PMCID: PMC10512299 DOI: 10.3389/ffunb.2021.736093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/20/2021] [Indexed: 09/26/2023]
Abstract
Fungal diseases such as candidiasis are some of the deadliest diseases among immunocompromised patients. These fungi naturally exist on human skin and throughout the digestive system. When the microbiota balance becomes upset, these fungi become pathogenic and potentially lethal. At the pathogenesis of fungal diseases, host immune system response is diverse. At the early stages of fungal pathogenesis such as Candida albicans, it was shown that these fungi use the immune cells of the host body and cause malfunction the early induction of proinflammatory cytokines of the host body leading to a reduction in their numbers. However, at some stages of fungal diseases, the immune response is severe. Despite many treatments already being available, it seems that one of the best treatments could be an immune-stimulatory agent. Some of the subsets of MSCs and exosome-derived cells, as a cell-to-cell communicator agent, have many roles in the human body, including anti-inflammatory and immune-modulatory effects. However, the TLR4-primed and IL-17+ subsets of MSCs have been shown to have immune-stimulatory effects. These subsets of the MSCs produce pro-inflammatory cytokines and reduce immunosuppressive cytokines and chemokines. Thus, they could trigger inflammation and stop fungal pathogenesis. As some biological activities and molecules inherit elements of their exosomes from their maternal cells, the exosome-derived TLR4-primed and IL-17+ subsets of MSCs could be a good candidate for fighting against fungal diseases. The applications of exosomes in human diseases are well-known and expanding. It is time to investigate the exosomes application in fungal diseases. In this review, the probable role of exosomes in treating fungal diseases is explored.
Collapse
|
205
|
Fazaeli H, Kalhor N, Naserpour L, Davoodi F, Sheykhhasan M, Hosseini SKE, Rabiei M, Sheikholeslami A. A Comparative Study on the Effect of Exosomes Secreted by Mesenchymal Stem Cells Derived from Adipose and Bone Marrow Tissues in the Treatment of Osteoarthritis-Induced Mouse Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9688138. [PMID: 34616850 PMCID: PMC8490078 DOI: 10.1155/2021/9688138] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Exosomes as extracellular vesicles (EVs) are nanoscale intercellular messengers secreted from cells to deliver biological signals. Today, exosomes have become a new field of research in regenerative medicine and are considered as potential therapies to control inflammation and wound healing and enhance and improve healing in many diseases. Given the global burden of osteoarthritis (OA) as the fastest-growing health condition and one of the major causes of physical disability in the aging population, research to establish EVs as therapeutic products can meet the basic clinical needs in the management of osteoarthritis and provide a therapeutic solution. OBJECTIVES The present study is aimed at evaluating the regenerative potentials of the exosomes secreted from adipose and bone marrow tissue-derived mesenchymal stem cells (AD- and BM-MSCs) in ameliorating the symptoms of OA. METHOD In this experimental study, AD- and BM-MSCs were isolated and cultured in the laboratory until passage 3. Finally, these cells' secreted exosomes were isolated from their conditioned medium. Ciprofloxacin-induced OA mouse models underwent intra-articular injection of exosomes from AD-MSCs and BM-MSCs. Finally, the expression levels of collagen I and II, sox9, and aggrecan genes using real-time PCR, histological analysis, and immunohistochemical (IHC) studies were performed. RESULTS Real-time PCR data showed that although the expression level of collagen type II was lower in both exosome-treated groups than the normal, but it was significantly increased in comparison with the sham and OA, with higher expression in BM-Exo rather than AD-Exo group. Similarly, the histological staining and IHC results have provided almost identical data, emphasizing on better therapeutic effect of BM-MSCs-exosome than AD-MSCs-exosome. CONCLUSION BM-MSCs secreted exosomes in comparison with AD-MSCs could be considered as a better therapeutic option to improve osteoarthritis and exhibit potential as a disease-modifying osteoarthritis cell-free product.
Collapse
Affiliation(s)
- Hoda Fazaeli
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Leila Naserpour
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Faezeh Davoodi
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Mohsen Sheykhhasan
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | | | - Mohammad Rabiei
- Department of Biology, Faculty of Science, Azad Islamic University of Qom, Qom, Iran
| | - Azar Sheikholeslami
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| |
Collapse
|
206
|
Stem Cells in Autologous Microfragmented Adipose Tissue: Current Perspectives in Osteoarthritis Disease. Int J Mol Sci 2021; 22:ijms221910197. [PMID: 34638538 PMCID: PMC8508703 DOI: 10.3390/ijms221910197] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a chronic debilitating disorder causing pain and gradual degeneration of weight-bearing joints with detrimental effects on cartilage volume as well as cartilage damage, generating inflammation in the joint structure. The etiology of OA is multifactorial. Currently, therapies are mainly addressing the physical and occupational aspects of osteoarthritis using pharmacologic pain treatment and/or surgery to manage the symptomatology of the disease with no specific regard to disease progression or prevention. Herein, we highlight alternative therapeutics for OA specifically considering innovative and encouraging translational methods with the use of adipose mesenchymal stem cells.
Collapse
|
207
|
Feng ZY, Yang SD, Wang T, Guo S. Effect of Melatonin for Regulating Mesenchymal Stromal Cells and Derived Extracellular Vesicles. Front Cell Dev Biol 2021; 9:717913. [PMID: 34540834 PMCID: PMC8440901 DOI: 10.3389/fcell.2021.717913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Melatonin is a hormone, synthesized in the pineal gland, which primarily controls the circadian rhythm of the body. In recent years, melatonin has also been shown to regulate metabolism, provide neuroprotection, and act as an anti-inflammatory, free radical scavenger. There has also been a recent research interest in the role of melatonin in regulating mesenchymal stromal cells (MSCs). MSCs are pivotal for their ability to differentiate into a variety of different tissues. There is also increasing evidence for the therapeutic prospects of MSCs via paracrine signaling. In addition to secreting cytokines and chemokines, MSCs can secrete extracellular vesicles (EVs), allowing them to respond to injury and promote tissue regeneration. While there has been a major research interest in the use of MSCs for regenerative medicine, the clinical application is limited by many risks, including tumorigenicity, senescence, and sensitivity to toxic environments. The use of MSC-derived EVs for cell-free therapy can potentially avoid the disadvantages of MSCs, which makes this an exciting prospect for regenerative medicine. Prior research has shown that MSCs, via paracrine mechanisms, can identify receptor-independent responses to melatonin and then activate a series of downstream pathways, which exert a variety of effects, including anti-tumor and anti-inflammatory effects. Here we review the synthesis of melatonin, its mechanisms of action, and the effect of melatonin on MSCs via paracrine signaling. Furthermore, we summarize the current clinical applications of melatonin and discuss future prospects.
Collapse
Affiliation(s)
- Zi-Yi Feng
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shu-De Yang
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ting Wang
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
208
|
Extracellular vesicles as novel approaches for the treatment of osteoarthritis: a narrative review on potential mechanisms. J Mol Histol 2021; 52:879-891. [PMID: 34510315 DOI: 10.1007/s10735-021-10017-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA) is a progressive degeneration of articular cartilage with involvement of synovial membrane, and subchondral bone. Current treatment approaches have focused on controlling the OA symptoms, pain, and inflammation. Recently, cell-based therapies, including the application of stem cells such as mesenchymal stem cells (MSCs), have been introduced for restoration of the articular cartilage. Despite promising outcomes, there are some limitations in the application of MSCs for OA treatment. It has been demonstrated that the regenerative potential of stem cells is related to the production of paracrine factors. Extracellular vehicles (EVs), the main component of cell secretome, are membrane-bounded structures that deliver biologically active agents. The delivery of molecules (e.g., nucleic acids, proteins, and lipids) leads to cell-to-cell communication and the alteration of cell functions. In this review, general characteristics of EVs, as well as their potential mechanisms in the prevention and treatment of OA were considered. Based on in vitro and in vivo studies, EVs have shown to contribute to cartilage regeneration via suppression of degenerative factors and regulation of chondrocyte function in the synthesis of extracellular matrix components. Also, they inhibit the progression of OA or protect the cartilage from degradation via their impact on inflammatory cytokines. The different signaling pathways of EVs against the pathologic features of OA were summarized in this review. According to the results obtained from several investigations, more investigations should be design to prove the safety and effectiveness of EVs in the treatment and prevention of OA progression.
Collapse
|
209
|
Mollentze J, Durandt C, Pepper MS. An In Vitro and In Vivo Comparison of Osteogenic Differentiation of Human Mesenchymal Stromal/Stem Cells. Stem Cells Int 2021; 2021:9919361. [PMID: 34539793 PMCID: PMC8443361 DOI: 10.1155/2021/9919361] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/23/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
The use of stem cells in regenerative medicine, including tissue engineering and transplantation, has generated a great deal of enthusiasm. Mesenchymal stromal/stem cells (MSCs) can be isolated from various tissues, most commonly, bone marrow but more recently adipose tissue, dental pulp, and Wharton's jelly, to name a few. MSCs display varying phenotypic profiles and osteogenic differentiating capacity depending and their site of origin. MSCs have been successfully differentiated into osteoblasts both in vitro an in vivo but discrepancies exist when the two are compared: what happens in vitro does not necessarily happen in vivo, and it is therefore important to understand why these differences occur. The osteogenic process is a complex network of transcription factors, stimulators, inhibitors, proteins, etc., and in vivo experiments are helpful in evaluating the various aspects of this osteogenic process without distractions and confounding variables. With that in mind, the results of in vitro experiments need to be carefully considered and interpreted with caution as they do not perfectly replicate the conditions found within living organisms. This is where in vivo experiments help us better understand interactions that might occur in the osteogenic process that cannot be replicated in vitro. Potentially, these differences could also be exploited to develop an optimal MSC cell therapeutic product that can be used for bone disorders. There are many bone disorders, most of which cause a great deal of discomfort. Clinically acceptable protocols could be developed in which MSCs are used to aid in bone regeneration providing relief for patients with chronic pain. The aim of this review is to examine the differences between studies conducted in vitro and in vivo with regard to the osteogenic process to better define the gaps in current osteogenic research. By better understanding osteogenic differentiation, we can better define treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Jamie Mollentze
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
210
|
Li Z, Huang Z, Bai L. Cell Interplay in Osteoarthritis. Front Cell Dev Biol 2021; 9:720477. [PMID: 34414194 PMCID: PMC8369508 DOI: 10.3389/fcell.2021.720477] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic disease and a significant health concern that needs to be urgently solved. OA affects the cartilage and entire joint tissues, including the subchondral bone, synovium, and infrapatellar fat pads. The physiological and pathological changes in these tissues affect the occurrence and development of OA. Understanding complex crosstalk among different joint tissues and their roles in OA initiation and progression is critical in elucidating the pathogenic mechanism of OA. In this review, we begin with an overview of the role of chondrocytes, synovial cells (synovial fibroblasts and macrophages), mast cells, osteoblasts, osteoclasts, various stem cells, and engineered cells (induced pluripotent stem cells) in OA pathogenesis. Then, we discuss the various mechanisms by which these cells communicate, including paracrine signaling, local microenvironment, co-culture, extracellular vesicles (exosomes), and cell tissue engineering. We particularly focus on the therapeutic potential and clinical applications of stem cell-derived extracellular vesicles, which serve as modulators of cell-to-cell communication, in the field of regenerative medicine, such as cartilage repair. Finally, the challenges and limitations related to exosome-based treatment for OA are discussed. This article provides a comprehensive summary of key cells that might be targets of future therapies for OA.
Collapse
Affiliation(s)
- Zihao Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyu Huang
- Foreign Languages College, Shanghai Normal University, Shanghai, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
211
|
Lin J, Wang L, Lin J, Liu Q. The Role of Extracellular Vesicles in the Pathogenesis, Diagnosis, and Treatment of Osteoarthritis. Molecules 2021; 26:4987. [PMID: 34443573 PMCID: PMC8398019 DOI: 10.3390/molecules26164987] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/10/2021] [Accepted: 08/14/2021] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects the entire joint and has been a tremendous burden on the health care system worldwide. Although cell therapy has made significant progress in the treatment of OA and cartilage regeneration, there are still a series of problems. Recently, more and more evidence shows that extracellular vesicles (EVs) play an important role in the progression and treatment of OA. Here, we discuss that EVs from different cell sources not only participate in OA progression, but can also be used as effective tools for the diagnosis and treatment of OA. In addition, cell pretreatment strategies and EV tissue engineering play an increasingly prominent role in the field of OA treatment. This article will systematically review the latest developments in these areas. As stated above, it may provide new insights for improving OA and cartilage regeneration.
Collapse
Affiliation(s)
- Jianjing Lin
- Arthritis Clinical and Research Center, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (J.L.); (J.L.)
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Li Wang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China;
| | - Jianhao Lin
- Arthritis Clinical and Research Center, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (J.L.); (J.L.)
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Qiang Liu
- Arthritis Clinical and Research Center, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (J.L.); (J.L.)
- Arthritis Institute, Peking University, Beijing 100044, China
| |
Collapse
|
212
|
Maiborodin I, Shevela A, Matveeva V, Morozov V, Toder M, Krasil’nikov S, Koryakina A, Shevela A, Yanushevich O. First Experimental Study of the Influence of Extracellular Vesicles Derived from Multipotent Stromal Cells on Osseointegration of Dental Implants. Int J Mol Sci 2021; 22:ijms22168774. [PMID: 34445482 PMCID: PMC8395855 DOI: 10.3390/ijms22168774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023] Open
Abstract
Herein, the aim was to study the state of the bone tissue adjacent to dental implants after the use of extracellular vesicles derived from multipotent stromal cells (MSC EVs) of bone marrow origin in the experiment. In compliance with the rules of asepsis and antiseptics under general intravenous anesthesia with propofol, the screw dental implants were installed in the proximal condyles of the tibia of outbred rabbits without and with preliminary introduction of 19.2 μg MSC EVs into each bone tissue defect. In 3, 7, and 10 days after the operation, the density of bone tissue adjacent to different parts of the implant using an X-ray unit with densitometer was measured. In addition, the histological examinations of the bone site with the hole from the removed device and the soft tissues from the surface of the proximal tibial condyle in the area of intra-bone implants were made. It was found out that 3 days after implantation with the use of MSC EVs, the bone density was statistically significantly higher by 47.2% than after the same implantation, but without the injection of MSC EVs. It is possible that as a result of the immunomodulatory action of MSC EVs, the activity of inflammation decreases, and, respectively, the degree of vasodilation in bones and leukocyte infiltration of the soft tissues are lower, in comparison with the surgery performed in the control group. The bone fragments formed during implantation are mainly consolidated with each other and with the regenerating bone. Day 10 demonstrated that all animals with the use of MSC EVs had almost complete fusion of the screw device with the bone tissue, whereas after the operation without the application of MSC EVs, the heterogeneous histologic pattern was observed: From almost complete osseointegration of the implant to the absolute absence of contact between the foreign body and the new formed bone. Therefore, the use of MSC EVs during the introduction of dental implants into the proximal condyle of the tibia of rabbits contributes to an increase of the bone tissue density near the device after 3 days and to the achievement of consistently successful osseointegration of implants 10 days after the surgery.
Collapse
Affiliation(s)
- Igor Maiborodin
- Laboratory of Health Management Technologies, The Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine, The Russian Academy of Sciences, Siberian Branch, Akademika Lavrenteva Str., 8, 630090 Novosibirsk, Russia; (A.S.); (V.M.); (V.M.); (S.K.); (A.S.)
- Institute of Molecular Pathology and Pathomorphology, Federal State Budget Scientific Institution “Federal Research Center of Fundamental and Translational Medicine” of the Ministry of Science and Higher Education of the Russian Federation, Akademika Timakova Str., 2, 630117 Novosibirsk, Russia
- Correspondence:
| | - Aleksandr Shevela
- Laboratory of Health Management Technologies, The Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine, The Russian Academy of Sciences, Siberian Branch, Akademika Lavrenteva Str., 8, 630090 Novosibirsk, Russia; (A.S.); (V.M.); (V.M.); (S.K.); (A.S.)
- International Center of Dental Implantology iDent, Sibrevkoma Str., 9b, 630007 Novosibirsk, Russia; (M.T.); (A.K.)
| | - Vera Matveeva
- Laboratory of Health Management Technologies, The Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine, The Russian Academy of Sciences, Siberian Branch, Akademika Lavrenteva Str., 8, 630090 Novosibirsk, Russia; (A.S.); (V.M.); (V.M.); (S.K.); (A.S.)
| | - Vitaly Morozov
- Laboratory of Health Management Technologies, The Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine, The Russian Academy of Sciences, Siberian Branch, Akademika Lavrenteva Str., 8, 630090 Novosibirsk, Russia; (A.S.); (V.M.); (V.M.); (S.K.); (A.S.)
| | - Michael Toder
- International Center of Dental Implantology iDent, Sibrevkoma Str., 9b, 630007 Novosibirsk, Russia; (M.T.); (A.K.)
| | - Sergey Krasil’nikov
- Laboratory of Health Management Technologies, The Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine, The Russian Academy of Sciences, Siberian Branch, Akademika Lavrenteva Str., 8, 630090 Novosibirsk, Russia; (A.S.); (V.M.); (V.M.); (S.K.); (A.S.)
| | - Alina Koryakina
- International Center of Dental Implantology iDent, Sibrevkoma Str., 9b, 630007 Novosibirsk, Russia; (M.T.); (A.K.)
| | - Andrew Shevela
- Laboratory of Health Management Technologies, The Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine, The Russian Academy of Sciences, Siberian Branch, Akademika Lavrenteva Str., 8, 630090 Novosibirsk, Russia; (A.S.); (V.M.); (V.M.); (S.K.); (A.S.)
| | - Oleg Yanushevich
- Moscow State University of Medicine and Dentistry, The Ministry of Healthcare of the Russian Federation, Delegatskaya Str., 20, p. 1, 127473 Moscow, Russia;
| |
Collapse
|
213
|
Kresakova L, Danko J, Vdoviakova K, Medvecky L, Zert Z, Petrovova E, Varga M, Spakovska T, Pribula J, Gasparek M, Giretova M, Stulajterova R, Kolvek F, Andrejcakova Z, Simaiova V, Kadasi M, Vrabec V, Toth T, Hura V. In Vivo Study of Osteochondral Defect Regeneration Using Innovative Composite Calcium Phosphate Biocement in a Sheep Model. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4471. [PMID: 34442993 PMCID: PMC8398687 DOI: 10.3390/ma14164471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 11/17/2022]
Abstract
This study aimed to clarify the therapeutic effect and regenerative potential of the novel, amino acids-enriched acellular biocement (CAL) based on calcium phosphate on osteochondral defects in sheep. Eighteen sheep were divided into three groups, the treated group (osteochondral defects filled with a CAL biomaterial), the treated group with a biocement without amino acids (C cement), and the untreated group (spontaneous healing). Cartilages of all three groups were compared with natural cartilage (negative control). After six months, sheep were evaluated by gross appearance, histological staining, immunohistochemical staining, histological scores, X-ray, micro-CT, and MRI. Treatment of osteochondral defects by CAL resulted in efficient articular cartilage regeneration, with a predominant structural and histological characteristic of hyaline cartilage, contrary to fibrocartilage, fibrous tissue or disordered mixed tissue on untreated defect (p < 0.001, modified O'Driscoll score). MRI results of treated defects showed well-integrated and regenerated cartilage with similar signal intensity, regularity of the articular surface, and cartilage thickness with respect to adjacent native cartilage. We have demonstrated that the use of new biocement represents an effective solution for the successful treatment of osteochondral defects in a sheep animal model, can induce an endogenous regeneration of cartilage in situ, and provides several benefits for the design of future therapies supporting osteochondral defect healing.
Collapse
Affiliation(s)
- Lenka Kresakova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (J.D.); (K.V.); (E.P.); (V.S.)
| | - Jan Danko
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (J.D.); (K.V.); (E.P.); (V.S.)
| | - Katarina Vdoviakova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (J.D.); (K.V.); (E.P.); (V.S.)
| | - Lubomir Medvecky
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (L.M.); (M.G.); (R.S.)
| | - Zdenek Zert
- Clinic of Horses, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.Z.); (F.K.); (V.H.)
| | - Eva Petrovova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (J.D.); (K.V.); (E.P.); (V.S.)
| | - Maros Varga
- Hospital AGEL Kosice-Saca, Lucna 57, 040 15 Kosice-Saca, Slovakia; (M.V.); (T.S.); (J.P.)
| | - Tatiana Spakovska
- Hospital AGEL Kosice-Saca, Lucna 57, 040 15 Kosice-Saca, Slovakia; (M.V.); (T.S.); (J.P.)
| | - Jozef Pribula
- Hospital AGEL Kosice-Saca, Lucna 57, 040 15 Kosice-Saca, Slovakia; (M.V.); (T.S.); (J.P.)
| | - Miroslav Gasparek
- Department of Engineering Science, University of Oxford, Parks Road, Oxford OX1 3PJ, UK;
| | - Maria Giretova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (L.M.); (M.G.); (R.S.)
| | - Radoslava Stulajterova
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, Watsonova 47, 040 01 Kosice, Slovakia; (L.M.); (M.G.); (R.S.)
| | - Filip Kolvek
- Clinic of Horses, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.Z.); (F.K.); (V.H.)
| | - Zuzana Andrejcakova
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Veronika Simaiova
- Department of Morphological Disciplines, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (J.D.); (K.V.); (E.P.); (V.S.)
| | - Marian Kadasi
- Clinic of Ruminants, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Vladimir Vrabec
- Clinic of Birds, Exotic and Free Living Animals, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Teodor Toth
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Kosice, Letna 9, 042 00 Kosice, Slovakia;
| | - Vladimir Hura
- Clinic of Horses, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 041 81 Kosice, Slovakia; (Z.Z.); (F.K.); (V.H.)
| |
Collapse
|
214
|
Jayaraman S, Gnanasampanthapandian D, Rajasingh J, Palaniyandi K. Stem Cell-Derived Exosomes Potential Therapeutic Roles in Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:723236. [PMID: 34447796 PMCID: PMC8382889 DOI: 10.3389/fcvm.2021.723236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Owing to myocardial abnormalities, cardiac ailments are considered to be the major cause of morbidity and mortality worldwide. According to a recent study, membranous vesicles that are produced naturally, termed as "exosomes", have emerged as the potential candidate in the field of cardiac regenerative medicine. A wide spectrum of stem cells has also been investigated in the treatment of cardiovascular diseases (CVD). Exosomes obtained from the stem cells are found to be cardioprotective and offer great hope in the treatment of CVD. The basic nature of exosomes is to deal with the intracellular delivery of both proteins and nucleic acids. This activity of exosomes helps us to rely on them as the attractive pharmaceutical delivery agents. Most importantly, exosomes derived from microRNAs (miRNAs) hold great promise in assessing the risk of CVD, as they serve as notable biomarkers of the disease. Exosomes are small, less immunogenic, and lack toxicity. These nanovesicles harbor immense potential as a therapeutic entity and would provide fruitful benefits if consequential research were focused on their upbringing and development as a useful diagnostic and therapeutic tool in the field of medicine.
Collapse
Affiliation(s)
- Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| | - Johnson Rajasingh
- Department of Bioscience Research & Medicine-Cardiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| |
Collapse
|
215
|
He Y, Ji D, Lu W, Chen G. The Mechanistic Effects and Clinical Applications of Various Derived Mesenchymal Stem Cells in Immune Thrombocytopenia. Acta Haematol 2021; 145:9-17. [PMID: 34515042 DOI: 10.1159/000517989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/22/2021] [Indexed: 12/16/2022]
Abstract
Immune thrombocytopenia (ITP) is an acquired autoimmune disorder characterized by persistent thrombocytopenia resulting from increased platelet destruction and a loss of autoimmune tolerance. The pathogenesis of ITP is highly complex. Although ITP may be effectively controlled with currently available medications in some patients, a subset of cases remain refractory. The application of mesenchymal stem cells (MSCs) for human hematopoietic stem cell transplantation has increasingly demonstrated that MSCs modulate innate or adaptive immunity, thus resulting in a tolerant microenvironment. Functional defects and immunomodulatory disorders have been observed after the use of bone marrow mesenchymal stem cells (BM-MSCs) from patients with ITP. Here, we summarize the underlying mechanisms and clinical applications of various derived MSCs for ITP treatment, focusing on the main mechanisms underlying the functional defects and immune dysfunction of BM-MSCs from patients with ITP. Functional effects associated with the activation of the p53 pathway include decreased activity of the phosphatidylinositol 3 kinase/Akt pathway and activation of the TNFAIP3/NF-κB/SMAD7 pathway. Immune dysfunction appears to be associated with an impaired ability of BM-MSCs to induce various types of immune cells in ITP. At present, research focusing on MSCs in ITP remains in preliminary stages. The application of autologous or exogenous MSCs in the clinical treatment of ITP has been attempted in only a small case study and must be validated in larger-scale clinical trials.
Collapse
Affiliation(s)
- Yue He
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dexiang Ji
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Lu
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guoan Chen
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
216
|
Qiao K, Chen Q, Cao Y, Li J, Xu G, Liu J, Cui X, Tian K, Zhang W. Diagnostic and Therapeutic Role of Extracellular Vesicles in Articular Cartilage Lesions and Degenerative Joint Diseases. Front Bioeng Biotechnol 2021; 9:698614. [PMID: 34422779 PMCID: PMC8371972 DOI: 10.3389/fbioe.2021.698614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/28/2021] [Indexed: 01/15/2023] Open
Abstract
Two leading contributors to the global disability are cartilage lesions and degenerative joint diseases, which are characterized by the progressive cartilage destruction. Current clinical treatments often fail due to variable outcomes and an unsatisfactory long-term repair. Cell-based therapies were once considered as an effective solution because of their anti-inflammatory and immunosuppression characteristics as well as their differentiation capacity to regenerate the damaged tissue. However, stem cell-based therapies have inherent limitations, such as a high tumorigenicity risk, a low retention, and an engraftment rate, as well as strict regulatory requirements, which result in an underwhelming therapeutic effect. Therefore, the non-stem cell-based therapy has gained its popularity in recent years. Extracellular vesicles (EVs), in particular, like the paracrine factors secreted by stem cells, have been proven to play a role in mediating the biological functions of target cells, and can achieve the therapeutic effect similar to stem cells in cartilage tissue engineering. Therefore, a comprehensive review of the therapeutic role of EVs in cartilage lesions and degenerative joint diseases can be discussed both in terms of time and favorability. In this review, we summarized the physiological environment of a joint and its pathological alteration after trauma and consequent changes in EVs, which are lacking in the current literature studies. In addition, we covered the potential working mechanism of EVs in the repair of the cartilage and the joint and also discussed the potential therapeutic applications of EVs in future clinical use.
Collapse
Affiliation(s)
- Kai Qiao
- First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Qi Chen
- First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yiguo Cao
- First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jie Li
- First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Gang Xu
- First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jiaqing Liu
- Qingdao University of Science and Technology, Qingdao, China
| | - Xiaolin Cui
- First Affiliated Hospital, Dalian Medical University, Dalian, China
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - Kang Tian
- First Affiliated Hospital, Dalian Medical University, Dalian, China
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Weiguo Zhang
- First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
217
|
Boulestreau J, Maumus M, Jorgensen C, Noël D. Extracellular vesicles from mesenchymal stromal cells: Therapeutic perspectives for targeting senescence in osteoarthritis. Adv Drug Deliv Rev 2021; 175:113836. [PMID: 34166759 DOI: 10.1016/j.addr.2021.113836] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 06/12/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is a common age-related disease that correlates with a high number of senescent cells in joint tissues. Senescence has been reported to be one of the main drivers of OA pathogenesis, in particular via the release of senescence-associated secretory phenotype (SASP) factors. SASP factors are secreted as single molecules and/or packaged within extracellular vesicles (EVs), thereby contributing to senescent phenotype dissemination. Targeting senescent cells using senolytics or senomorphics has therefore been tested and improvement of OA-associated features has been reported in murine models. Mesenchymal stromal cells (MSCs) and their derived EVs (MSC-EVs) are promising treatments for OA, exerting pleiotropic functions by producing a variety of factors. However, functions of MSCs and MSC-EVs are affected by aging. In this review, we discuss on the impact of the senescent environment on functions of aged MSC-EVs and on the anti-aging properties of MSC-EVs in the context of OA.
Collapse
Affiliation(s)
| | - Marie Maumus
- IRMB, University of Montpellier, INSERM, Montpellier, France; Bauerfeind France, IRMB, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, France.
| |
Collapse
|
218
|
Hade MD, Suire CN, Suo Z. Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells 2021; 10:1959. [PMID: 34440728 PMCID: PMC8393426 DOI: 10.3390/cells10081959] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a type of extracellular vesicles, produced within multivesicular bodies, that are then released into the extracellular space through a merging of the multivesicular body with the plasma membrane. These vesicles are secreted by almost all cell types to aid in a vast array of cellular functions, including intercellular communication, cell differentiation and proliferation, angiogenesis, stress response, and immune signaling. This ability to contribute to several distinct processes is due to the complexity of exosomes, as they carry a multitude of signaling moieties, including proteins, lipids, cell surface receptors, enzymes, cytokines, transcription factors, and nucleic acids. The favorable biological properties of exosomes including biocompatibility, stability, low toxicity, and proficient exchange of molecular cargos make exosomes prime candidates for tissue engineering and regenerative medicine. Exploring the functions and molecular payloads of exosomes can facilitate tissue regeneration therapies and provide mechanistic insight into paracrine modulation of cellular activities. In this review, we summarize the current knowledge of exosome biogenesis, composition, and isolation methods. We also discuss emerging healing properties of exosomes and exosomal cargos, such as microRNAs, in brain injuries, cardiovascular disease, and COVID-19 amongst others. Overall, this review highlights the burgeoning roles and potential applications of exosomes in regenerative medicine.
Collapse
Affiliation(s)
| | | | - Zucai Suo
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA; (M.D.H.); (C.N.S.)
| |
Collapse
|
219
|
Park S, Bello A, Arai Y, Ahn J, Kim D, Cha KY, Baek I, Park H, Lee SH. Functional Duality of Chondrocyte Hypertrophy and Biomedical Application Trends in Osteoarthritis. Pharmaceutics 2021; 13:pharmaceutics13081139. [PMID: 34452101 PMCID: PMC8400409 DOI: 10.3390/pharmaceutics13081139] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Chondrocyte hypertrophy is one of the key indicators in the progression of osteoarthritis (OA). However, compared with other OA indications, such as cartilage collapse, sclerosis, inflammation, and protease activation, the mechanisms by which chondrocyte hypertrophy contributes to OA remain elusive. As the pathological processes in the OA cartilage microenvironment, such as the alterations in the extracellular matrix, are initiated and dictated by the physiological state of the chondrocytes, in-depth knowledge of chondrocyte hypertrophy is necessary to enhance our understanding of the disease pathology and develop therapeutic agents. Chondrocyte hypertrophy is a factor that induces OA progression; it is also a crucial factor in the endochondral ossification. This review elaborates on this dual functionality of chondrocyte hypertrophy in OA progression and endochondral ossification through a description of the characteristics of various genes and signaling, their mechanism, and their distinguishable physiological effects. Chondrocyte hypertrophy in OA progression leads to a decrease in chondrogenic genes and destruction of cartilage tissue. However, in endochondral ossification, it represents an intermediate stage at the process of differentiation of chondrocytes into osteogenic cells. In addition, this review describes the current therapeutic strategies and their mechanisms, involving genes, proteins, cytokines, small molecules, three-dimensional environments, or exosomes, against the OA induced by chondrocyte hypertrophy. Finally, this review proposes that the contrasting roles of chondrocyte hypertrophy are essential for both OA progression and endochondral ossification, and that this cellular process may be targeted to develop OA therapeutics.
Collapse
Affiliation(s)
- Sunghyun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea
| | - Alvin Bello
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Jinsung Ahn
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Dohyun Kim
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Kyung-Yup Cha
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Inho Baek
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Hansoo Park
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Correspondence: ; Tel.: +82-31-961-5153; Fax: +82-31-961-5108
| |
Collapse
|
220
|
Bei HP, Hung PM, Yeung HL, Wang S, Zhao X. Bone-a-Petite: Engineering Exosomes towards Bone, Osteochondral, and Cartilage Repair. SMALL 2021; 17:e2101741. [PMID: 34288410 DOI: 10.1002/smll.202101741] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/18/2021] [Indexed: 02/05/2023]
Abstract
Recovery from bone, osteochondral, and cartilage injuries/diseases has been burdensome owing to the damaged vasculature of large defects and/or avascular nature of cartilage leading to a lack of nutrients and supplying cells. However, traditional means of treatment such as microfractures and cell-based therapy only display limited efficacy due to the inability to ensure cell survival and potential aggravation of surrounding tissues. Exosomes have recently emerged as a powerful tool for this tissue repair with its complex content of transcription factors, proteins, and targeting ligands, as well as its unique ability to home in on target cells thanks to its phospholipidic nature. They are engineered to serve specialized applications including enhancing repair, anti-inflammation, regulating homeostasis, etc. via means of physical, chemical, and biological modulations in its deriving cell culture environments. This review focuses on the engineering means to functionalize exosomes for bone, osteochondral, and cartilage regeneration, with an emphasis on conditions such as osteoarthritis, osteoporosis, and osteonecrosis. Finally, future implications for exosome development will be given alongside its potential combination with other strategies to improve its therapeutic efficacy in the osteochondral niche.
Collapse
Affiliation(s)
- Ho Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Pak Ming Hung
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Hau Lam Yeung
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Rd, Pokfulam, Hong Kong SAR, 999077, China
| | - Shuqi Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.,Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, 610065, China.,Institute for Advanced Study, Chengdu University, Chengdu, 610106, P. R. China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| |
Collapse
|
221
|
Li Z, Li M, Xu P, Ma J, Zhang R. Compositional Variation and Functional Mechanism of Exosomes in the Articular Microenvironment in Knee Osteoarthritis. Cell Transplant 2021; 29:963689720968495. [PMID: 33086893 PMCID: PMC7784575 DOI: 10.1177/0963689720968495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a major cause of disability worldwide with increasing age. Knee OA (KOA) is the most prevalent type of OA. Recently, it is considered that KOA is a whole joint disease, including articular cartilage, subchondral bone, synovium, ligaments, joint capsules, and muscles around the joint. Exosomes in knee joint are mainly secreted by articular chondrocytes and synoviocytes. They participate in cell and tissue cross-talk by carrying a complex cargo of proteins, lipids, nucleic acids, etc. Under normal conditions, exosomes maintain the microenvironmental homeostasis of the joint cavity. Under pathological conditions, the composition and function of exosomes changes, which in turn, disrupts the balance of anabolism and catabolism of articular chondrocyte and facilitates inflammatory responses, thus accelerating KOA progression. As a regenerative medicine, mesenchymal stem cells (MSCs) are promised to facilitate repair of degenerated cartilage and decelerate OA process. The therapeutic function of MSC mainly depends on MSC-derived exosomes, which can restore the homeostasis of the articular microenvironment. In the future, the specific mechanism of exosomes for OA treatment needs further elucidation, and the treatment effect of exosomes for long-term and/or severe OA needs further exploration.
Collapse
Affiliation(s)
- Zheng Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.,Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Manling Li
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jie Ma
- Medical Research Center, Xi'an No. 3 Hospital, Xi'an, China.,School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Rui Zhang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
222
|
Zhang Q, Shen Y, Zhao S, Jiang Y, Zhou D, Zhang Y. Exosomes miR-15a promotes nucleus pulposus-mesenchymal stem cells chondrogenic differentiation by targeting MMP-3. Cell Signal 2021; 86:110083. [PMID: 34252537 DOI: 10.1016/j.cellsig.2021.110083] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 06/29/2021] [Accepted: 07/07/2021] [Indexed: 12/27/2022]
Abstract
The physiology of the nucleus pulposus (NP) in intervertebral disc degeneration (IVD) has been studied widely. However, interactions involving nucleus pulposus -mesenchymal stem cells (NP-MSCs) are less understood. MicroRNA 15a (miR-15a) is known to target and modulate genes involved in cellular proliferation and apoptosis. This study aimed to understand the interactions and impact of miR-15a and NP-MSCs on chondrogenic differentiation and IVD degeneration. Exosomes secreted by NP cells were purified by differential centrifugation and identified by transmission electron microscopy and exosomal markers. Further, by co-culture these exosomes were re-introduced into the NP-MSC cells, which were confirmed by fluorescence confocal microscopy. NP-MSCs treated with exo-miR-15a increases aggrecan and collagen II mRNA and protein levels while decreasing mRNA and protein levels of ADAMTS4/5 and MMP-3/-13. Toluidine blue staining confirmed that chondrogenic differentiation was increased in NP-MSCs treated with exo-miR-15a. NP-MSCs treated with exo-anti-miR-15a inhibit aggrecan and collagen II expression while increasing ADAMTS4/5 and MMP-3/-13 expression and decreasing chondrogenic differentiation. Dual-luciferase reporter assays revealed that miR-15a directly targets MMP-3 and downregulates its expression. Overexpression of miR-15a increased proliferation and colony formation, whereas combinatorial overexpression with MMP3, suppressed miR-15a's effects. This was also evident through the decreased phosphorylation of PI3K and Akt, upregulation of Wnt3a and β-catenin in the presence of miR-15a, but overexpression of MMP3 indicated an opposite effect. Overall, these data demonstrate that exo-miR-15a promotes NP-MSCs chondrogenic differentiation by downregulating MMP-3 through PI3K/Akt and Wnt3a/β-catenin axis.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Yifei Shen
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Shujie Zhao
- Department of Orthopedics, The People's Hospital of Jiangsu Province, Nanjing, Jiangsu 210029, China
| | - Yuqing Jiang
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Dong Zhou
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, Jiangsu 213003, China.
| | - Yunkun Zhang
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, Jiangsu 213003, China.
| |
Collapse
|
223
|
Ghafouri-Fard S, Niazi V, Hussen BM, Omrani MD, Taheri M, Basiri A. The Emerging Role of Exosomes in the Treatment of Human Disorders With a Special Focus on Mesenchymal Stem Cells-Derived Exosomes. Front Cell Dev Biol 2021; 9:653296. [PMID: 34307345 PMCID: PMC8293617 DOI: 10.3389/fcell.2021.653296] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are produced by diverse eukaryotic and prokaryotic cells. They have prominent roles in the modulation of cell-cell communication, inflammation versus immunomodulation, carcinogenic processes, cell proliferation and differentiation, and tissue regeneration. These acellular vesicles are more promising than cellular methods because of the lower risk of tumor formation, autoimmune responses and toxic effects compared with cell therapy. Moreover, the small size and lower complexity of these vesicles compared with cells have made their production and storage easier than cellular methods. Exosomes originated from mesenchymal stem cells has also been introduced as therapeutic option for a number of human diseases. The current review aims at summarization of the role of EVs in the regenerative medicine with a focus on their therapeutic impacts in liver fibrosis, lung disorders, osteoarthritis, colitis, myocardial injury, spinal cord injury and retinal injury.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
224
|
Yang Y, Zhu Z, Gao R, Yuan J, Zhang J, Li H, Xie Z, Wang Y. Controlled release of MSC-derived small extracellular vesicles by an injectable Diels-Alder crosslinked hyaluronic acid/PEG hydrogel for osteoarthritis improvement. Acta Biomater 2021; 128:163-174. [PMID: 33862283 DOI: 10.1016/j.actbio.2021.04.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) show great therapeutic potential for osteoarthritis (OA). However, their low bioavailability through intraarticular injection inhibits the process of clinical application. In the present study, an injectable Diels-Alder crosslinked hyaluronic acid/PEG (DAHP) hydrogel was developed as an intraarticular delivery platform for MSC-sEVs. Our results showed that the DAHP hydrogel could be prepared easily and that its gelation properties were suitable for intraarticular administration. In vitro studies demonstrated that the DAHP hydrogel could achieve sustained release of MSC-sEVs mainly by degradation control and preserve the therapeutic functions of sEVs. An in vivo experiment revealed that the DAHP hydrogel could enhance the efficacy of MSC-sEVs for OA improvement. This study provides a suitable delivery platform for MSC-sEVs-based OA therapy. STATEMENT OF SIGNIFICANCE: Mesenchymal stem cell (MSC)-derived small extracellular vesicles (MSC-sEVs) have shown a high potential as a cell-free therapeutic factor for treating osteoarthritis (OA). The sustained release of these MSC-sEVs in the joint space is essential for their clinical application. Herein, an injectable Diels-Alder crosslinked hyaluronic acid/PEG (DAHP) hydrogel was developed for intraarticular release of MSC-sEVs. The properties of the DAHP hydrogel, namely gelation features, cytocompatibility, sustained release, and functional maintenance of MSC-sEVs, make it suitable for intraarticular injection and delivery of sEVs. The efficacy of MSC-sEVs was enhanced by the intraarticularly injected DAHP hydrogel. Our present study provides a promising sustained delivery platform for MSC-sEVs for treating OA.
Collapse
Affiliation(s)
- Yunlong Yang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Zhaochen Zhu
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China; Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Renzhi Gao
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China; Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Ji Yuan
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Juntao Zhang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Haiyan Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800#, Dongchuan Road, Shanghai 200240, China; Chemical and Environmental Engineering, School of Engineering, RMIT University, 124 La Trobe St, Melbourne VIC 3000, Australia
| | - Zongping Xie
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China.
| | - Yang Wang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| |
Collapse
|
225
|
Ke Z, Zhu J. Stem-Cell Derived Exosomes for the Treatment of Osteoarthritis. Curr Stem Cell Res Ther 2021; 15:597-601. [PMID: 32026786 DOI: 10.2174/1574888x15666200206104332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/02/2019] [Accepted: 11/29/2019] [Indexed: 01/08/2023]
Abstract
Osteoarthritis(OA) is a common degenerative orthopedic disease with multiple pathologic changes in joints affecting large populations worldwide. No treatment can reverse the progress of OA. Since exosomes were first reported in 1983, researches have been conducted to explore the mechanisms and therapeutic potential of exosomes in treating OA. Exosomes derived from Mesenchymal stem cells have attracted increasing attention in tackling the disease. This article summarizes the current advances and challenges in exosomes for OA, which may providea reference for further research.
Collapse
Affiliation(s)
- Zekai Ke
- Department of Orthopaedics, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, Guangdong Province 518055, China
| | - Jinyu Zhu
- Department of Orthopaedics, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, Guangdong Province 518055, China
| |
Collapse
|
226
|
Li S, Liu J, Liu S, Jiao W, Wang X. Mesenchymal stem cell-derived extracellular vesicles prevent the development of osteoarthritis via the circHIPK3/miR-124-3p/MYH9 axis. J Nanobiotechnology 2021; 19:194. [PMID: 34193158 PMCID: PMC8244143 DOI: 10.1186/s12951-021-00940-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 06/20/2021] [Indexed: 12/16/2022] Open
Abstract
Background Extracellular vesicles (EVs) secreted by mesenchymal stem cells (MSCs) may play a vital role in a variety of biological processes, including cartilage regeneration. However, few studies reported their potential in the development of osteoarthritis (OA) previously. In this study, we explored the biological roles and underlying mechanism of MSCs-EVs in OA. Results Co-culture experiments revealed that MSCs-EVs could promote the expression of collagen type II alpha 1 chain (COL2A1), SRY-box transcription factor 9 (SOX9) and Aggrecan while negatively regulate the expression of chondrocyte hypertrophy markers matrix metallopeptidase 13 (MMP-13) and RUNX family transcription factor 2 (Runx2) in mouse chondrocytes in the OA model. Besides, the results of cell experiments indicated that MSCs-EVs could notably weaken the suppression of chondrocyte proliferation, migration and the promotion of chondrocyte apoptosis via interleukin1β (IL-1β) induction. In addition, MSCs-circHIPK3-EVs (EVs derived from MSCs overexpressing circHIPK3) considerably improved IL-1β-induced chondrocyte injury. Mechanistically, we elucidated that circHIPK3 could directly bind to miR-124-3p and subsequently elevate the expression of the target gene MYH9. Conclusion The findings in our study demonstrated that EVs-circHIPK3 participated in MSCs-EVs-mediated chondrocyte proliferation and migration induction and in chondrocyte apoptosis inhibition via the miR-124-3p/MYH9 axis. This offers a promising novel cell-free therapy for treating OA. Graphic abstract ![]()
Collapse
Affiliation(s)
- Shenglong Li
- Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.,Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning Province, China
| | - Jie Liu
- Department of Prosthodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, China
| | - Siyu Liu
- Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Weijie Jiao
- Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Xiaohong Wang
- Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China. .,Center of Organ Manufacturing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
227
|
An artificial membrane binding protein-polymer surfactant nanocomplex facilitates stem cell adhesion to the cartilage extracellular matrix. Biomaterials 2021; 276:120996. [PMID: 34280823 DOI: 10.1016/j.biomaterials.2021.120996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/12/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
One of the major challenges within the emerging field of injectable stem cell therapies for articular cartilage (AC) repair is the retention of sufficient viable cell numbers at the site of injury. Even when delivered via intra-articular injection, the number of stem cells retained at the target is often low and declines rapidly over time. To address this challenge, an artificial plasma membrane binding nanocomplex was rationally designed to provide human mesenchymal stem cells (hMSCs) with increased adhesion to articular cartilage tissue. The nanocomplex comprises the extracellular matrix (ECM) binding peptide of a placenta growth factor-2 (PlGF-2) fused to a supercharged green fluorescent protein (scGFP), which was electrostatically conjugated to anionic polymer surfactant chains to yield [S-]scGFP_PlGF2. The [S-]scGFP_PlGF2 nanocomplex spontaneously inserts into the plasma membrane of hMSCs, is not cytotoxic, and does not inhibit differentiation. The nanocomplex-modified hMSCs showed a significant increase in affinity for immobilised collagen II, a key ECM protein of cartilage, in both static and dynamic cell adhesion assays. Moreover, the cells adhered strongly to bovine ex vivo articular cartilage explants resulting in high cell numbers. These findings suggest that the re-engineering of hMSC membranes with [S-]scGFP_PlGF2 could improve the efficacy of injectable stem cell-based therapies for the treatment of damaged articular cartilage.
Collapse
|
228
|
Ćulum NM, Cooper TT, Bell GI, Hess DA, Lagugné-Labarthet F. Characterization of extracellular vesicles derived from mesenchymal stromal cells by surface-enhanced Raman spectroscopy. Anal Bioanal Chem 2021; 413:5013-5024. [PMID: 34137912 DOI: 10.1007/s00216-021-03464-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are secreted by all cells into bodily fluids and play an important role in intercellular communication through the transfer of proteins and RNA. There is evidence that EVs specifically released from mesenchymal stromal cells (MSCs) are potent cell-free regenerative agents. However, for MSC EVs to be used in therapeutic practices, there must be a standardized and reproducible method for their characterization. The detection and characterization of EVs are a challenge due to their nanoscale size as well as their molecular heterogeneity. To address this challenge, we have fabricated gold nanohole arrays of varying sizes and shapes by electron beam lithography. These platforms have the dual purpose of trapping single EVs and enhancing their vibrational signature in surface-enhanced Raman spectroscopy (SERS). In this paper, we report SERS spectra for MSC EVs derived from pancreatic tissue (Panc-MSC) and bone marrow (BM-MSC). Using principal component analysis (PCA), we determined that the main compositional differences between these two groups are found at 1236, 761, and 1528 cm-1, corresponding to amide III, tryptophan, and an in-plane -C=C- vibration, respectively. We additionally explored several machine learning approaches to distinguish between BM- and Panc-MSC EVs and achieved 89 % accuracy, 89 % sensitivity, and 88 % specificity using logistic regression.
Collapse
Affiliation(s)
- Nina M Ćulum
- Department of Chemistry, Centre for Advanced Materials and Biomaterials Research (CAMBR), University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - Tyler T Cooper
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of medicine and Dentistry, University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - Gillian I Bell
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of medicine and Dentistry, University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - David A Hess
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of medicine and Dentistry, University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - François Lagugné-Labarthet
- Department of Chemistry, Centre for Advanced Materials and Biomaterials Research (CAMBR), University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
229
|
Zhang B, Tian X, Hao J, Xu G, Zhang W. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Tissue Regeneration. Cell Transplant 2021; 29:963689720908500. [PMID: 32207341 PMCID: PMC7444208 DOI: 10.1177/0963689720908500] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells that have attracted
increasing interest in the field of regenerative medicine. Previously, the
differentiation ability of MSCs was believed to be primarily responsible for
tissue repair. Recent studies have shown that paracrine mechanisms play an
important role in this process. MSCs can secrete soluble molecules and
extracellular vesicles (EVs), which mediate paracrine communication. EVs contain
large amounts of proteins and nucleic acids, such as mRNAs and microRNAs
(miRNAs), and can transfer the cargo between cells. The cargoes are similar to
those in MSCs and are not susceptible to degradation due to the protection of
the EV bimolecular membrane structure. MSC-EVs can mimic the biological
characteristics of MSCs, such as differentiation, maturation, and self-renewal.
Due to their broad biological functions and their ability to transfer molecules
between cells, EVs have been intensively studied by an increasing number of
researchers with a focus on therapeutic applications, especially those of EVs
secreted by MSCs. In this review, we discuss MSC-derived EVs and their
therapeutic potential in tissue regeneration.
Collapse
Affiliation(s)
- Bocheng Zhang
- Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, Chin.,Dalian Medical University, Dalian, Liaoning, China
| | | | - Jun Hao
- Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, Chin
| | - Gang Xu
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases, Liaoning, China
| | - Weiguo Zhang
- Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, Chin
| |
Collapse
|
230
|
Liao Q, Li BJ, Li Y, Xiao Y, Zeng H, Liu JM, Yuan LX, Liu G. Low-intensity pulsed ultrasound promotes osteoarthritic cartilage regeneration by BMSC-derived exosomes via modulating the NF-κB signaling pathway. Int Immunopharmacol 2021; 97:107824. [PMID: 34102487 DOI: 10.1016/j.intimp.2021.107824] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022]
Abstract
Osteoarthritis is the most common disabling joint disease throughout the world, and the effect of therapy on its course is still unsatisfactory in clinical practice. Recent studies have shown that mesenchymal stem cell (MSC)-derived exosomes can promote cartilage repair and regeneration in osteoarthritis, indicating that these exosomes could be a novel and promising strategy for treating osteoarthritis. This study investigated whether low-intensity pulsed ultrasound (LIPUS) enhances the effects of bone marrow MSC (BMSC)-derived exosomes on cartilage regeneration in osteoarthritis and examined the underlying mechanism. Our results revealed that BMSC-derived exosomes display the typical morphological features of exosomes. LIPUS-mediated BMSC-derived exosomes promoted cartilage regeneration, increased chondrocyte proliferation and extracellular matrix synthesis, suppressed inflammation, and inhibited the interleukin (IL)-1β-induced activation of the nuclear factor kappa B (NF-κB) pathway. In brief, LIPUS enhances the promoting effects of BMSC-derived exosomes on osteoarthritic cartilage regeneration, mainly by strengthening the inhibition of inflammation and further enhancing chondrocyte proliferation and cartilage matrix synthesis. The underlying mechanism could be related to the inhibition of the IL-1β-induced activation of the NF-κB pathway.
Collapse
Affiliation(s)
- Qing Liao
- Department of Rehabilitation Medicine, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China; Department of Rehabilitation Medicine, Shunde Hospital of Southern Medical University, Foshan 528000, China
| | - Bao Jian Li
- Department of Rehabilitation Medicine, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Yang Li
- Department of Rehabilitation Medicine, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Yu Xiao
- Department of Rehabilitation Medicine, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Hui Zeng
- Department of Rehabilitation Medicine, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Jie Mei Liu
- Department of Rehabilitation Medicine, Shunde Hospital of Southern Medical University, Foshan 528000, China
| | - Li Xia Yuan
- Southern Medical University, Guangzhou 510000, China.
| | - Gang Liu
- Department of Rehabilitation Medicine, Shunde Hospital of Southern Medical University, Foshan 528000, China; Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou 510000, China.
| |
Collapse
|
231
|
Yao Z, Li J, Xiong H, Cui H, Ning J, Wang S, Ouyang X, Qian Y, Fan C. MicroRNA engineered umbilical cord stem cell-derived exosomes direct tendon regeneration by mTOR signaling. J Nanobiotechnology 2021; 19:169. [PMID: 34090456 PMCID: PMC8180131 DOI: 10.1186/s12951-021-00906-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/21/2021] [Indexed: 12/27/2022] Open
Abstract
Background Exosomes are extracellular vesicles of nano-structures and represent an emerging nano-scale acellular therapy in recent years. Tendon regeneration is a sophisticated process in the field of microsurgery due to its poor natural healing ability. To date, no successful long-term solution has been provided for the healing of tendon injuries. Functional recovery requires advanced treatment strategies. Human umbilical cord mesenchymal stem cell-derived exosomes (HUMSC-Exos) are considered as promising cell-free therapeutic agents. However, few studies reported their potential in the tendon repair previously. In this study, we explored the roles and underlying mechanisms of HUMSC-Exos in the tendon regeneration. Results Expression of tendon‐specific markers in, and collagen deposition by, tendon-derived stem cells (TDSCs) treated with HUMSC-Exos increased in vitro. In a rat Achilles tendon injury model, treatment with HUMSC-Exos improved the histological structure, enhanced tendon-specific matrix components, and optimized biomechanical properties of the Achilles tendon. Findings in miRNA sequencing indicated a significant increase in miR-29a-3p in HUMSC-Exo-treated Achilles tendons. Next, luciferase assay in combination with western blot identified phosphatase and tensin homolog (PTEN) as the specific target of miR-29a-3p. Furthermore, we applied a miR-29a-3p-specific agonist to engineer HUMSC-Exos. These HUMSC-Exos overexpressing miR-29a-3p amplified the gain effects of HUMSC-Exos on tendon healing in vivo. To explore the underlying mechanisms, a transforming growth factor-β1 (TGF-β1) inhibitor (SB-431542), mTOR inhibitor (rapamycin), and engineered HUMSC-Exos were employed. The results showed that TGF-β1 and mTOR signaling were involved in the beneficial effects of HUMSC-Exos on tendon regeneration. Conclusion The findings in our study suggest that PTEN/mTOR/TGF-β1 signaling cascades may be a potential pathway for HUMSC-Exos to deliver miR-29a-3p for tendon healing and implicate a novel therapeutic strategy for tendon regeneration via engineered stem cell-derived exosomes. Graphic abstract ![]()
Collapse
Affiliation(s)
- Zhixiao Yao
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hao Xiong
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Haomin Cui
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jiexin Ning
- Department of Plastics, Binzhou People's Hospital, Binzhou, 256610, China
| | - Shikun Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xingyu Ouyang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yun Qian
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
232
|
Qin B, Zhang Q, Chen D, Yu HY, Luo AX, Suo LP, Cai Y, Cai DY, Luo J, Huang JF, Xiong K. Extracellular vesicles derived from mesenchymal stem cells: A platform that can be engineered. Histol Histopathol 2021; 36:615-632. [PMID: 33398872 DOI: 10.14670/hh-18-297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells play an important role in tissue damage and repair. This role is mainly due to a paracrine mechanism, and extracellular vesicles (EVs) are an important part of the paracrine function. EVs play a vital role in many aspects of cell homeostasis, physiology, and pathology, and EVs can be used as clinical biomarkers, vaccines, or drug delivery vehicles. A large number of studies have shown that EVs derived from mesenchymal stem cells (MSC-EVs) play an important role in the treatment of various diseases. However, the problems of low production, low retention rate, and poor targeting of MSC-EVs are obstacles to current clinical applications. The engineering transformation of MSC-EVs can make up for those shortcomings, thereby improving treatment efficiency. This review summarizes the latest research progress of MSC-EV direct and indirect engineering transformation from the aspects of improving MSC-EV retention rate, yield, targeting, and MSC-EV visualization research, and proposes some feasible MSC-EV engineering methods of transformation.
Collapse
Affiliation(s)
- Bo Qin
- Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Qi Zhang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Dan Chen
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hai-Yang Yu
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ai-Xiang Luo
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Liang-Peng Suo
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yan Cai
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - De-Yang Cai
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jia Luo
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ju-Fang Huang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China.
| | - Kun Xiong
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China.
| |
Collapse
|
233
|
Zhang B, Tian X, Qu Z, Liu J, Yang L, Zhang W. Efficacy of extracellular vesicles from mesenchymal stem cells on osteoarthritis in animal models: a systematic review and meta-analysis. Nanomedicine (Lond) 2021; 16:1297-1310. [PMID: 34044578 DOI: 10.2217/nnm-2021-0047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: Some studies have reported results from the use of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) to treat osteoarthritis (OA). Objective: To evaluate the efficacy of MSC-EVs as a treatment for OA. Data sources: Databases were searched using the terms 'mesenchymal stem cells', 'osteoarthritis' and 'extracellular vesicles.' Study eligibility criteria: Studies performed in animal models utilizing MSC-EVs to treat OA that described the macroscopic evaluation or histological evaluation were included. Study appraisal: The quality of the studies was examined using the CAMARADES quality checklist. Results: MSC-EVs were superior to the placebo in the macroscopic evaluation and histological evaluation. MSC-EVs were more effective in the early stage of OA and once a week was better than multiple times a week. Limitations: The included studies were highly heterogeneous. Conclusion: MSC-EVs may improve the results of macroscopic and histological evaluations of OA.
Collapse
Affiliation(s)
- Bocheng Zhang
- Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China.,Graduate School, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Xiaoyuan Tian
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China.,Graduate School, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Zhenan Qu
- Orthopedics IV, Affiliated Zhongshan Hospital of Dalian University, Liaoning, 116000, China
| | - Jiaming Liu
- Graduate School, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Liang Yang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Weiguo Zhang
- Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China.,Graduate School, Dalian Medical University, Dalian, Liaoning, 116000, China
| |
Collapse
|
234
|
Extracellular Vesicles from Mesenchymal Stem Cells as Potential Treatments for Osteoarthritis. Cells 2021; 10:cells10061287. [PMID: 34067325 PMCID: PMC8224601 DOI: 10.3390/cells10061287] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disorder of the joint and its prevalence and severity is increasing owing to ageing of the population. Osteoarthritis is characterized by the degradation of articular cartilage and remodeling of the underlying bone. There is little understanding of the cellular and molecular processes involved in pathophysiology of OA. Currently the treatment for OA is limited to painkillers and anti-inflammatory drugs, which only treat the symptoms. Some patients may also undergo surgical procedures to replace the damaged joints. Extracellular vesicles (EV) play an important role in intercellular communications and their concentration is elevated in the joints of OA patients, although their mechanism is unclear. Extracellular vesicles are naturally released by cells and they carry their origin cell information to be delivered to target cells. On the other hand, mesenchymal stem cells (MSCs) are highly proliferative and have a great potential in cartilage regeneration. In this review, we provide an overview of the current OA treatments and their limitations. We also discuss the role of EV in OA pathophysiology. Finally, we highlight the therapeutic potential of MSC-derived EV in OA and their challenges.
Collapse
|
235
|
Miao C, Zhou W, Wang X, Fang J. The Research Progress of Exosomes in Osteoarthritis, With Particular Emphasis on the Mediating Roles of miRNAs and lncRNAs. Front Pharmacol 2021; 12:685623. [PMID: 34093208 PMCID: PMC8176107 DOI: 10.3389/fphar.2021.685623] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a kind of degenerative disease, which is caused by many factors such as aging, obesity, strain, trauma, congenital joint abnormalities, joint deformities. Exosomes are mainly derived from the invagination of intracellular lysosomes, which are released into the extracellular matrix after fusion of the outer membrane of multi vesicles with the cell membrane. Exosomes mediate intercellular communication and regulate the biological activity of receptor cells by carrying non-coding RNA, long noncoding RNAs (lncRNAs), microRNAs (miRNAs), proteins and lipids. Evidences show that exosomes are involved in the pathogenesis of OA. In view of the important roles of exosomes in OA, this paper systematically reviewed the roles of exosomes in the pathogenesis of OA, including the roles of exosomes in OA diagnosis, the regulatory mechanisms of exosomes in the pathogenesis, and the intervention roles of exosomes in the treatment of OA. Reviewing the roles of exosomes in OA will help to clarify the pathogenesis of OA and explore new diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Department of Pharmacy, School of Life and Health Sciences, Anhui University of Science and Technology, Fengyang, China.,Institute of Prevention and Treatment of Rheumatoid Arthritis of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Wanwan Zhou
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Jihong Fang
- Department of Nursing, Anhui Provincial Children's Hospital, Affiliated to Anhui Medical University, Hefei, China.,Department of Orthopedics, Anhui Provincial Children's Hospital, Affiliated to Anhui Medical University, Hefei, China
| |
Collapse
|
236
|
Huang J, Xiong J, Yang L, Zhang J, Sun S, Liang Y. Cell-free exosome-laden scaffolds for tissue repair. NANOSCALE 2021; 13:8740-8750. [PMID: 33969373 DOI: 10.1039/d1nr01314a] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
With the development of regenerative medicine, tissue repair at the molecular, cellular, tissue, and organ level has seen continuous improvements over traditional techniques. As the core of tissue repair, seed cells are widely used in various fields of regenerative medicine. However, their use is still associated with problems such as decreased cell survival and regeneration capacity after transplantation, immune rejection, and ethical concerns. Therefore, it is difficult to universally and safely apply stem cell banks for regenerative medicine. The paracrine effects of cells, especially secretion of exosomes, play vital roles in cell communication, immune response, angiogenesis, scar formation, tissue repair, and other biological functions. Exosomes are a type of nanoscale extracellular vesicle that contain biologically active molecules such as RNA and proteins; therefore, exosomes can replicate the functions of their parental cells. Meanwhile, exosomes can be used as nanocarriers to deliver active factors or small molecules to promote tissue repair. Preclinical studies of exosomes in tissue engineering and regenerative medicine have been carried in the fields of bone/cartilage repair, nerve regeneration, liver and kidney regeneration, skin repair, vascular tissue regeneration, etc. This review introduces exosomes from the aspects of biogenesis, composition, identification, and isolation, and focuses on the development status of scaffold materials for exosome delivery. In addition, we highlight examples of exosome-laden scaffolds for preclinical applications in tissue repair. We look forward to the broad application prospects of exosome-laden scaffolds.
Collapse
Affiliation(s)
- Jianghong Huang
- Department of Orthopedics, Shenzhen Second People's Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, China and Tsinghua University Shenzhen International Graduate School, Innovation Leading Engineering Doctor, Class 9 of 2020, Shenzhen, 518055, China
| | - Jianyi Xiong
- Department of Orthopedics, Shenzhen Second People's Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, China
| | - Lei Yang
- Department of Orthopedics, Shenzhen Second People's Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, China
| | - Jun Zhang
- Tsinghua University Shenzhen International Graduate School, Innovation Leading Engineering Doctor, Class 9 of 2020, Shenzhen, 518055, China
| | - Shuqing Sun
- Tsinghua University Shenzhen International Graduate School, Institute of Biomedicine and Health Engineering, Shenzhen, 518055, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen Key Laboratory for Psychological Healthcare & Shenzhen Institute of Mental Health, Shenzhen, 518020, China.
| |
Collapse
|
237
|
He F, Li L, Fan R, Wang X, Chen X, Xu Y. Extracellular Vesicles: An Emerging Regenerative Treatment for Oral Disease. Front Cell Dev Biol 2021; 9:669011. [PMID: 34079801 PMCID: PMC8165191 DOI: 10.3389/fcell.2021.669011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular Vesicles (EVs) are small lipid-enclosed particles containing biological molecules such as RNA and proteins that have emerged as vital modulators of intercellular communication. Increasingly, studies have shown that EVs play an essential role in the occurrence and prognosis of oral diseases. EVs are increasingly considered a research hotspot of oral diseases. In addition, the characteristics of carrying active molecules have also been studied in oral tissue regeneration. Evidence has shown that EVs regulate the homeostasis of the inflammatory microenvironment, promote angiogenesis, and repair damaged tissues. In this review, we summarized the characteristics of EVs and highlighted the role of EVs in oral tissue regeneration, including dental pulp, periodontal tissue, cartilage, and bone. We also discussed their deficiencies and prospects as a potential therapeutic role in the regeneration treatment of oral disease.
Collapse
Affiliation(s)
- Fanzhen He
- Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Lu Li
- Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ruyi Fan
- Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoqian Wang
- Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yan Xu
- Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
238
|
Li D, Gupta P, Sgaglione NA, Grande DA. Exosomes Derived from Non-Classic Sources for Treatment of Post-Traumatic Osteoarthritis and Cartilage Injury of the Knee: In Vivo Review. J Clin Med 2021; 10:jcm10092001. [PMID: 34066986 PMCID: PMC8124969 DOI: 10.3390/jcm10092001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis of the knee is one of the most common chronic, debilitating musculoskeletal conditions. Current conservative treatment modalities such as weight loss, non-steroidal anti-inflammatory drugs, and intra-articular steroid injections often only provide temporary pain relief and are unsatisfactory for long-term management. Though end stage osteoarthritis of the knee can be managed with total knee arthroplasty (TKA), finding alternative non-surgical options to delay or prevent the need for TKA are needed due to the increased healthcare costs and expenditures associated with TKA. Exosomes have been of particular interest given recent findings highlighting that stem cells may at least partially mediate some of their effects through the release of extracellular vesicles, such as exosomes. As such, better understanding the biological mechanisms and potential therapeutic effects of these exosomes is necessary. Here, we review in vivo studies that highlight the potential clinical use of exosomes derived from non-classical sources (not bone marrow or adipose derived MSCs derived MSCs) for osteoarthritis of the knee.
Collapse
Affiliation(s)
- Dan Li
- Orthopedic Research Laboratory, The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, NY 11030, USA;
| | - Puneet Gupta
- Department of Orthopaedic Surgery, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA;
| | - Nicholas A. Sgaglione
- Department of Orthopaedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA;
| | - Daniel A. Grande
- Orthopedic Research Laboratory, The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, NY 11030, USA;
- Department of Orthopaedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA;
- Correspondence: ; Tel.: +1-(516)-562-1138
| |
Collapse
|
239
|
Brose TZ, Kubosch EJ, Schmal H, Stoddart MJ, Armiento AR. Crosstalk Between Mesenchymal Stromal Cells and Chondrocytes: The Hidden Therapeutic Potential for Cartilage Regeneration. Stem Cell Rev Rep 2021; 17:1647-1665. [PMID: 33954877 DOI: 10.1007/s12015-021-10170-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Cartilage injuries following trauma create a puzzling clinical scenario. The finite reparative potential of articular cartilage is well known, and injuries are associated with an increased risk of osteoarthritis. Cell-based therapies have spotlighted chondrocytes and mesenchymal stromal cells (MSCs) as the functional unit of articular cartilage and the progenitor cells, respectively. The available clinical treatments cannot reproduce the biomechanical properties of articular cartilage and call for continuous investigations into alternative approaches. Co-cultures of chondrocytes and MSCs are an attractive in vitro system to step closer to the in vivo multicellular environment's complexity. Research on the mechanisms of interaction between both cell types will reveal essential cues to understand cartilage regeneration. This review describes the latest discoveries on these interactions, along with advantages and main challenges in vitro and in vivo. The successful clinical translation of in vitro studies requires establishing rigorous standards and clinically relevant research models and an organ-targeting therapeutic strategy.
Collapse
Affiliation(s)
- Teresa Z Brose
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.,Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Eva J Kubosch
- Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Hagen Schmal
- Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.,Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Angela R Armiento
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.
| |
Collapse
|
240
|
Song H, Zhao J, Cheng J, Feng Z, Wang J, Momtazi-Borojeni AA, Liang Y. Extracellular Vesicles in chondrogenesis and Cartilage regeneration. J Cell Mol Med 2021; 25:4883-4892. [PMID: 33942981 PMCID: PMC8178250 DOI: 10.1111/jcmm.16290] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs), mainly exosomes and microvesicles, are bilayer lipids containing biologically active information, including nucleic acids and proteins. They are involved in cell communication and signalling, mediating many biological functions including cell growth, migration and proliferation. Recently, EVs have received great attention in the field of tissue engineering and regenerative medicine. Many in vivo and in vitro studies have attempted to evaluate the chondrogenesis potential of these microstructures and their roles in cartilage regeneration. EVs derived from mesenchymal stem cells (MSCs) or chondrocytes have been found to induce chondrocyte proliferation and chondrogenic differentiation of stem cells in vitro. Preclinical studies have shown that exosomes derived from MSCs have promising results in cartilage repair and in cell‐free therapy of osteoarthritis. This review will focus on the in vitro and in vivo chondrogenesis and cartilage regeneration of EVs as well as their potential in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Hong Song
- Department of Orthopedics, Guizhou Province Orthopedics Hospital, Guiyang, Guizhou, China
| | - Jiasong Zhao
- Department of International Ward, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Cheng
- Department of Spine Surgery, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Zhijie Feng
- Department of Geriatric Orthopaedics, Tangshan City Second Hospital, Hebei Province, Tangshan, China
| | - Jianhua Wang
- Department Bone Microsurgery, Sanya people's Hospital, Sanya, China
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yimin Liang
- Department of Orthopedics, Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Taizhou, China
| |
Collapse
|
241
|
Liu A, Lin D, Zhao H, Chen L, Cai B, Lin K, Shen SG. Optimized BMSC-derived osteoinductive exosomes immobilized in hierarchical scaffold via lyophilization for bone repair through Bmpr2/Acvr2b competitive receptor-activated Smad pathway. Biomaterials 2021; 272:120718. [PMID: 33838528 DOI: 10.1016/j.biomaterials.2021.120718] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cell-derived exosomes (MSC-exos), with its inherent capacity to modulate cellular behavior, are emerging as a novel cell-free therapy for bone regeneration. Herein, focusing on practical applying problems, the osteoinductivity of MSC-exos produced by different stem cell sources (rBMSCs/rASCs) and culture conditions (osteoinductive/common) were systematically compared to screen out an optimized osteogenic exosome (BMSC-OI-exo). Via bioinformatic analyses by miRNA microarray and in vitro pathway verification by gene silencing and miRNA transfection, we first revealed that the osteoinductivity of BMSC-OI-exo was attributed to multi-component exosomal miRNAs (let-7a-5p, let-7c-5p, miR-328a-5p and miR-31a-5p). These miRNAs targeted Acvr2b/Acvr1 and regulated the competitive balance of Bmpr2/Acvr2b toward Bmpr-elicited Smad1/5/9 phosphorylation. On these bases, lyophilized delivery of BMSC-OI-exo on hierarchical mesoporous bioactive glass (MBG) scaffold was developed to realize bioactivity maintenance and sustained release by entrapment in the surface microporosity of the scaffold. In a rat cranial defect model, the loading of BMSC-OI-exo efficiently enhanced the bone forming capacity of the scaffold and induced rapid initiation of bone regeneration. This paper could provide empirical bases of MSC-exo-based therapy for bone regeneration and theoretical bases of MSC-exo-induced osteogenesis mechanism. The BMSC-OI-exo-loaded MBG scaffold developed here represented a promising bone repairing strategy for future clinical application.
Collapse
Affiliation(s)
- Anqi Liu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, PR China
| | - Dan Lin
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, PR China.
| | - Hanjiang Zhao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, PR China
| | - Long Chen
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, PR China
| | - Bolei Cai
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, PR China; State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China.
| | - Kaili Lin
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, PR China.
| | - Steve Gf Shen
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, PR China; Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
242
|
Fatty Acids and Oxylipins in Osteoarthritis and Rheumatoid Arthritis-a Complex Field with Significant Potential for Future Treatments. Curr Rheumatol Rep 2021; 23:41. [PMID: 33913032 PMCID: PMC8081702 DOI: 10.1007/s11926-021-01007-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Purpose of Review Osteoarthritis (OA) and rheumatoid arthritis (RA) are characterized by abnormal lipid metabolism manifested as altered fatty acid (FA) profiles of synovial fluid and tissues and in the way dietary FA supplements can influence the symptoms of especially RA. In addition to classic eicosanoids, the potential roles of polyunsaturated FA (PUFA)-derived specialized pro-resolving lipid mediators (SPM) have become the focus of intensive research. Here, we summarize the current state of knowledge of the roles of FA and oxylipins in the degradation or protection of synovial joints. Recent Findings There exists discordance between the large body of literature from cell culture and animal experiments on the adverse and beneficial effects of individual FA and the lack of effective treatments for joint destruction in OA and RA patients. Saturated 16:0 and 18:0 induce mostly deleterious effects, while long-chain n-3 PUFA, especially 20:5n-3, have positive influence on joint health. The situation can be more complex for n-6 PUFA, such as 18:2n-6, 20:4n-6, and its derivative prostaglandin E2, with a combination of potentially adverse and beneficial effects. SPM analogs have future potential as analgesics for arthritic pain. Summary Alterations in FA profiles and their potential implications in SPM production may affect joint lubrication, synovial inflammation, pannus formation, as well as cartilage and bone degradation and contribute to the pathogeneses of inflammatory joint diseases. Further research directions include high-quality randomized controlled trials on dietary FA supplements and investigations on the significance of lipid composition of microvesicle membrane and cargo in joint diseases.
Collapse
|
243
|
Fan Y, Cui C, Li P, Bi R, Lyu P, Li Y, Zhu S. Fibrocartilage Stem Cells in the Temporomandibular Joint: Insights From Animal and Human Studies. Front Cell Dev Biol 2021; 9:665995. [PMID: 33987185 PMCID: PMC8111285 DOI: 10.3389/fcell.2021.665995] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/06/2021] [Indexed: 02/05/2023] Open
Abstract
Temporomandibular disorders (TMD) are diseases involving the temporomandibular joint (TMJ), masticatory muscles, and osseous components. TMD has a high prevalence, with an estimated 4.8% of the U.S. population experiencing signs and symptoms, and represents a financial burden to both individuals and society. During TMD progression, the most frequently affected site is the condylar cartilage. Comprising both fibrous and cartilaginous tissues, condylar cartilage has restricted cell numbers but lacks a vascular supply and has limited regenerative properties. In 2016, a novel stem cell niche containing a reservoir of fibrocartilage stem cells (FCSCs) was discovered in the condylar cartilage of rats. Subsequently, FCSCs were identified in mouse, rabbit, and human condylar cartilage. Unlike mesenchymal stem cells or other tissue-specific stem/progenitor cells, FCSCs play a unique role in the development and regeneration of fibrocartilage. More importantly, engraftment treatment of FCSCs has been successfully applied in animal models of TMD. In this context, FCSCs play a major role in the regeneration of newly formed cartilage. Furthermore, FCSCs participate in the regeneration of intramembranous bone by interacting with endothelial cells in bone defects. This evidence highlights the potential of FCSCs as an ideal stem cell source for the regeneration of oral maxillofacial tissue. This review is intended to detail the current knowledge of the characteristics and function of FCSCs in the TMJ, as well as the potential therapeutic applications of FCSCs. A deep understanding of the properties of FCSCs can thus inform the development of promising, biologically based strategies for TMD in the future.
Collapse
Affiliation(s)
- Yi Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chen Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Province Key Laboratory of Stomatology, Guangzhou, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ruiye Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Lyu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Songsong Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
244
|
Huang CY, Vesvoranan O, Yin X, Montoya A, Londono V, Sawatari Y, Garcia-Godoy F. Anti-Inflammatory Effects of Conditioned Medium of Periodontal Ligament-Derived Stem Cells on Chondrocytes, Synoviocytes, and Meniscus Cells. Stem Cells Dev 2021; 30:537-547. [PMID: 33757298 DOI: 10.1089/scd.2021.0010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis, afflicting millions of people in the world. Elevation of inflammatory mediators and enzymatic matrix destruction is often associated with OA. Therefore, the objective of this study was to investigate the effects of conditioned medium from periodontal ligament-derived stem cells (PDLSCs) on inflammatory and catabolic gene expressions of chondrocytes, synoviocytes, and meniscus cells under in vitro inflammatory condition. Stem cells were isolated from human periodontal ligaments. Conditioned medium was collected and concentrated 20 × . Chondrocytes, synoviocytes, and meniscus cells were isolated from pig knees and divided into four experimental groups: serum-free media, serum-free media+interleukin-1β (IL-1β) (10 ng/mL), conditioned media (CM), and CM+IL-1β. Protein content and extracellular vesicle (EV) miRNAs of CM were analyzed by liquid chromatography-tandem mass spectrometry and RNA sequencing, respectively. It was found that the IL-1β treatment upregulated the expression of IL-1β, tumor necrosis factor-α (TNF-α), MMP-13, and ADAMTS-4 genes in the three cell types, whereas PDLSC-conditioned medium prevented the upregulation of gene expression by IL-1β in all three cell types. This study also found that there was consistency in anti-inflammatory effects of PDLSC CM across donors and cell subcultures, while PDLSCs released several anti-inflammatory factors and EV miRNAs at high levels. OA has been suggested as an inflammatory disease in which all intrasynovial tissues are involved. PDLSC-conditioned medium is a cocktail of trophic factors and EV miRNAs that could mediate different inflammatory processes in various tissues in the joint. Introducing PDLSC-conditioned medium to osteoarthritic joints could be a potential treatment to prevent OA progression by inhibiting inflammation.
Collapse
Affiliation(s)
- Chun-Yuh Huang
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Oraya Vesvoranan
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Xue Yin
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Amanda Montoya
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Valeria Londono
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Yoh Sawatari
- Division of Oral and Maxillofacial Surgery, University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Franklin Garcia-Godoy
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
245
|
Velot É, Madry H, Venkatesan JK, Bianchi A, Cucchiarini M. Is Extracellular Vesicle-Based Therapy the Next Answer for Cartilage Regeneration? Front Bioeng Biotechnol 2021; 9:645039. [PMID: 33968913 PMCID: PMC8102683 DOI: 10.3389/fbioe.2021.645039] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/15/2021] [Indexed: 01/22/2023] Open
Abstract
"Extracellular vesicles" (EVs) is a term gathering biological particles released from cells that act as messengers for cell-to-cell communication. Like cells, EVs have a membrane with a lipid bilayer, but unlike these latter, they have no nucleus and consequently cannot replicate. Several EV subtypes (e.g., exosomes, microvesicles) are described in the literature. However, the remaining lack of consensus on their specific markers prevents sometimes the full knowledge of their biogenesis pathway, causing the authors to focus on their biological effects and not their origins. EV signals depend on their cargo, which can be naturally sourced or altered (e.g., cell engineering). The ability for regeneration of adult articular cartilage is limited because this avascular tissue is partly made of chondrocytes with a poor proliferation rate and migration capacity. Mesenchymal stem cells (MSCs) had been extensively used in numerous in vitro and preclinical animal models for cartilage regeneration, and it has been demonstrated that their therapeutic effects are due to paracrine mechanisms involving EVs. Hence, using MSC-derived EVs as cell-free therapy tools has become a new therapeutic approach to improve regenerative medicine. EV-based therapy seems to show similar cartilage regenerative potential compared with stem cell transplantation without the associated hindrances (e.g., chromosomal aberrations, immunogenicity). The aim of this short review is to take stock of occurring EV-based treatments for cartilage regeneration according to their healing effects. The article focuses on cartilage regeneration through various sources used to isolate EVs (mature or stem cells among others) and beneficial effects depending on cargos produced from natural or tuned EVs.
Collapse
Affiliation(s)
- Émilie Velot
- Faculté de Médecine, Biopôle de l’Université de Lorraine, Campus Brabois-Santé, Laboratoire UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Université de Lorraine, Vandoeuvre-Lès-Nancy, France
- Campus Brabois-Santé, Laboratoire de Travaux Pratiques de Physiologie, Faculté de Pharmacie, Université de Lorraine, Vandoeuvre-Lès-Nancy, France
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | | | - Arnaud Bianchi
- Campus Brabois-Santé, Laboratoire de Travaux Pratiques de Physiologie, Faculté de Pharmacie, Université de Lorraine, Vandoeuvre-Lès-Nancy, France
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| |
Collapse
|
246
|
Li Q, Yu H, Sun M, Yang P, Hu X, Ao Y, Cheng J. The tissue origin effect of extracellular vesicles on cartilage and bone regeneration. Acta Biomater 2021; 125:253-266. [PMID: 33657452 DOI: 10.1016/j.actbio.2021.02.039] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/22/2021] [Accepted: 02/24/2021] [Indexed: 12/30/2022]
Abstract
Direct implantation of mesenchymal stem cells (MSCs) for cartilage and bone tissue engineering faces challenges, such as immune rejection and loss of cellular viability or functionality. As nanoscale natural particles, exosomes or small extracellular vesicles (EVs) of MSCs have potential to circumvent these problems. It is significant to investigate the impact of the tissue origin of MSCs on the therapeutic bioactivity of their corresponding EVs for cartilage and bone regeneration. Here, rat MSCs isolated from the adipose, bone marrow, and synovium are cultured to obtain their corresponding EVs (ADSC-EVs, BMSC-EVs, and SMSC-EVs, respectively). The ADSC-EVs stimulate the migration, proliferation, and chondrogenic and osteogenic differentiation of BMSCs in vitro as well as cartilage and bone regeneration in a mouse model more than the BMSC-EVs or SMSC-EVs. Proteomics analysis reveals that the tissue origin contributes to the distinct protein profiles among the three types of EVs, which induced cartilage and bone regenerative capacities by potential mechanisms of regulating signaling pathways including focal adhesion, ECM-receptor interaction, actin cytoskeleton, cAMP, and PI3K-Akt signaling pathways. Consequently, these findings provide insight that the adipose may be a superior candidate in EV-based nanomedicine for cartilage and bone regeneration. STATEMENT OF SIGNIFICANCE: Extracelluar vesicles (EVs) of mesenchymal stem cells (MSCs) have been considered as a promising approach in cartilage and bone tissue engineering. In this study, for the first time, we investigated the tissue origin effect of EVs on chondrogenesis and osteogenesis of MSCs in vitro and in vivo. The results demonstrated that EVs of adipose-derived MSCs showed the most efficiency. Meanwhile, protein proteomics revealed the potential mechanisms. We provide a novel evidence that the adipose is a superior reservoir in EV-based nanotechnologies and biomaterials for cartilage and bone regeneration.
Collapse
Affiliation(s)
- Qi Li
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Huilei Yu
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Muyang Sun
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Peng Yang
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Yingfang Ao
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China.
| | - Jin Cheng
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China.
| |
Collapse
|
247
|
Chen Y, Tian Z, He L, Liu C, Wang N, Rong L, Liu B. Exosomes derived from miR-26a-modified MSCs promote axonal regeneration via the PTEN/AKT/mTOR pathway following spinal cord injury. Stem Cell Res Ther 2021; 12:224. [PMID: 33820561 PMCID: PMC8022427 DOI: 10.1186/s13287-021-02282-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/11/2021] [Indexed: 12/16/2022] Open
Abstract
Background Exosomes derived from the bone marrow mesenchymal stem cell (MSC) have shown great potential in spinal cord injury (SCI) treatment. This research was designed to investigate the therapeutic effects of miR-26a-modified MSC-derived exosomes (Exos-26a) following SCI. Methods Bioinformatics and data mining were performed to explore the role of miR-26a in SCI. Exosomes were isolated from miR-26a-modified MSC culture medium by ultracentrifugation. A series of experiments, including assessment of Basso, Beattie and Bresnahan scale, histological evaluation, motor-evoked potential recording, diffusion tensor imaging, and western blotting, were performed to determine the therapeutic influence and the underlying molecular mechanisms of Exos-26a in SCI rats. Results Exos-26a was shown to promote axonal regeneration. Furthermore, we found that exosomes derived from miR-26a-modified MSC could improve neurogenesis and attenuate glial scarring through PTEN/AKT/mTOR signaling cascades. Conclusions Exosomes derived from miR-26a-modified MSC could activate the PTEN-AKT-mTOR pathway to promote axonal regeneration and neurogenesis and attenuate glia scarring in SCI and thus present great potential for SCI treatment. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02282-0.
Collapse
Affiliation(s)
- Yuyong Chen
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Zhenming Tian
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Lei He
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Can Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Nangxiang Wang
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China
| | - Limin Rong
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.
| | - Bin Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
248
|
Kim GB, Shon OJ, Seo MS, Choi Y, Park WT, Lee GW. Mesenchymal Stem Cell-Derived Exosomes and Their Therapeutic Potential for Osteoarthritis. BIOLOGY 2021; 10:285. [PMID: 33915850 PMCID: PMC8066608 DOI: 10.3390/biology10040285] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/18/2022]
Abstract
Exosomes are nano-sized vesicles (50-150 nm in diameter) that contain nucleic acids (e.g., microRNA and messenger RNA), functional proteins, and bioactive lipids. They are secreted by various types of cells, including B cells, T cells, reticulocytes, dendritic cells, mast cells, epithelial cells, and mesenchymal stem cells (MSCs). They perform a wide variety of functions, including the repair of damaged tissues, regulation of immune responses, and reduction in inflammation. When considering the limitations of MSCs, including issues in standardization and immunogenicity, MSC-derived exosomes have advantages such as small dimensions, low immunogenicity, and lack of requirement for additional procedures for culture expansion or delivery. MSC-derived exosomes have shown outstanding therapeutic effects through chondro-protective and anti-inflammatory properties. MSC-derived exosomes may enable a new therapeutic paradigm for the treatment of osteoarthritis. However, further research is needed to prove their clinical effectiveness and feasibility.
Collapse
Affiliation(s)
- Gi Beom Kim
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University, Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea; (G.B.K.); (O.-J.S.); (W.T.P.)
| | - Oog-Jin Shon
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University, Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea; (G.B.K.); (O.-J.S.); (W.T.P.)
| | - Min-Soo Seo
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea;
| | - Young Choi
- Department of Orthopedic Surgery, Kosin University College of Medicine, Kosin University Gospel Hospital, 262 Gamcheon-ro, Seogu, Busan 49267, Korea;
| | - Wook Tae Park
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University, Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea; (G.B.K.); (O.-J.S.); (W.T.P.)
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University, Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea; (G.B.K.); (O.-J.S.); (W.T.P.)
| |
Collapse
|
249
|
Mustonen AM, Nieminen P. Extracellular Vesicles and Their Potential Significance in the Pathogenesis and Treatment of Osteoarthritis. Pharmaceuticals (Basel) 2021; 14:ph14040315. [PMID: 33915903 PMCID: PMC8065796 DOI: 10.3390/ph14040315] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by inflammation, gradual destruction of articular cartilage, joint pain, and functional limitations that eventually lead to disability. Join tissues, including synovium and articular cartilage, release extracellular vesicles (EVs) that have been proposed to sustain joint homeostasis as well as to contribute to OA pathogenesis. EVs transport biologically active molecules, and OA can be characterized by altered EV counts and composition in synovial fluid. Of EV cargo, specific non-coding RNAs could have future potential as diagnostic biomarkers for early OA. EVs may contribute to the propagation of inflammation and cartilage destruction by transporting and enhancing the production of inflammatory mediators and cartilage-degrading proteinases. In addition to inducing OA-related gene expression patterns in synoviocytes and articular chondrocytes, EVs can induce anti-OA effects, including increased extracellular matrix deposition and cartilage protection. Especially mesenchymal stem cell-derived EVs can alleviate intra-articular inflammation and relieve OA pain. In addition, surgically- or chemically-induced cartilage defects have been repaired with EV therapies in animal models. While human clinical trials are still in the future, the potential of actual cures to OA by EV products is very promising.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland;
- Department of Environmental and Biological Sciences, Faculty of Science and Forestry, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland
- Correspondence: ; Tel.: +358-294-45-1111
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland;
| |
Collapse
|
250
|
Murali VP, Holmes CA. Biomaterial-based extracellular vesicle delivery for therapeutic applications. Acta Biomater 2021; 124:88-107. [PMID: 33454381 DOI: 10.1016/j.actbio.2021.01.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicle (EV)- based therapies have been successfully tested in preclinical models for several biomedical applications, including tissue engineering, drug delivery and cancer therapy. However, EVs are most commonly delivered via local or systemic injection, which results in rapid clearance. In order to prolong the retention of EVs at target site and improve their therapeutic efficacy, biomaterial-based delivery systems are being investigated. This review discusses the various biomaterial-based systems that have been used to deliver EVs for therapeutic applications, specifically highlighting any strategies for controlled release. Further, challenges to clinical translation of biomaterial-based EV delivery systems are also discussed.
Collapse
Affiliation(s)
- Vishnu Priya Murali
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA.
| | - Christina A Holmes
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA.
| |
Collapse
|