301
|
Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. Int J Oral Sci 2019; 11:23. [PMID: 31423011 PMCID: PMC6802669 DOI: 10.1038/s41368-019-0060-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
In modern medicine, bone and dental loss and defects are common and widespread morbidities, for which regenerative therapy has shown great promise. Mesenchymal stem cells, obtained from various sources and playing an essential role in organ development and postnatal repair, have exhibited enormous potential for regenerating bone and dental tissue. Currently, mesenchymal stem cells (MSCs)-based bone and dental regeneration mainly includes two strategies: the rescue or mobilization of endogenous MSCs and the application of exogenous MSCs in cytotherapy or tissue engineering. Nevertheless, the efficacy of MSC-based regeneration is not always fulfilled, especially in diseased microenvironments. Specifically, the diseased microenvironment not only impairs the regenerative potential of resident MSCs but also controls the therapeutic efficacy of exogenous MSCs, both as donors and recipients. Accordingly, approaches targeting a diseased microenvironment have been established, including improving the diseased niche to restore endogenous MSCs, enhancing MSC resistance to a diseased microenvironment and renormalizing the microenvironment to guarantee MSC-mediated therapies. Moreover, the application of extracellular vesicles (EVs) as cell-free therapy has emerged as a promising therapeutic strategy. In this review, we summarize current knowledge regarding the tactics of MSC-based bone and dental regeneration and the decisive role of the microenvironment, emphasizing the therapeutic potential of microenvironment-targeting strategies in bone and dental regenerative medicine.
Collapse
|
302
|
The Role of Exosomes in Bone Remodeling: Implications for Bone Physiology and Disease. DISEASE MARKERS 2019; 2019:9417914. [PMID: 31485281 PMCID: PMC6710799 DOI: 10.1155/2019/9417914] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022]
Abstract
Bone remodeling represents a physiological phenomenon of continuous bone tissue renewal that requires fine orchestration of multiple cell types, which is critical for the understanding of bone disease but not yet clarified in precise detail. Exosomes, which are cell-secreted nanovesicles drawing increasing attention for their broad biosignaling functions, can shed new light on how multiple heterogeneous cells communicate for the purpose of bone remodeling. In the healthy bone, exosomes transmit signals favoring both bone synthesis and resorption, regulating the differentiation, recruitment, and activity of most cell types involved in bone remodeling and even assuming an active role in extracellular matrix mineralization. Additionally, in the ailing bone, they actively participate in pathogenic processes constituting also potential therapeutic agents and drug vectors. The present review summarizes the current knowledge on bone exosomes and bone remodeling in health and disease.
Collapse
|
303
|
Chen S, Tang Y, Liu Y, Zhang P, Lv L, Zhang X, Jia L, Zhou Y. Exosomes derived from miR-375-overexpressing human adipose mesenchymal stem cells promote bone regeneration. Cell Prolif 2019; 52:e12669. [PMID: 31380594 PMCID: PMC6797519 DOI: 10.1111/cpr.12669] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/29/2019] [Accepted: 05/04/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives The present study aimed to investigate whether exosomes derived from miR‐375‐overexpressing human adipose mesenchymal stem cells (hASCs) could enhance bone regeneration. Materials and Methods Exosomes enriched with miR‐375 (Exo [miR‐375]) were generated from hASCs stably overexpressing miR‐375 after lentiviral transfection and identified with transmission electron microscopy, nanosight and western blotting. The construction efficiency of Exo (miR‐375) was evaluated with qRT‐PCR and incubated with human bone marrow mesenchymal stem cells (hBMSCs) to optimize the effective dosage. Then, the osteogenic capability of Exo (miR‐375) was investigated with ALP and ARS assays. Furthermore, dual‐luciferase reporter assay and western blotting were conducted to reveal the underlying mechanism of miR‐375 in osteogenic regulation. Finally, Exo (miR‐375) were embedded with hydrogel and applied to a rat model of calvarial defect, and μ‐CT analysis and histological examination were conducted to evaluate the therapeutic effects of Exo (miR‐375) in bone regeneration. Results miR‐375 could be enriched in exosomes by overexpressing in the parent cells. Administration of Exo (miR‐375) at 50 μg/mL improved the osteogenic differentiation of hBMSCs. With miR‐375 absorbed by hBMSCs, insulin‐like growth factor binding protein 3 (IGFBP3) was inhibited by binding to its 3′UTR, and recombinant IGFBP3 protein reduced the osteogenic effects triggered by Exo (miR‐375). After incorporated with hydrogel, Exo (miR‐375) displayed a slow and controlled release, and further in vivo analysis demonstrated that Exo (miR‐375) enhanced the bone regenerative capacity in a rat model of calvarial defect. Conclusions Taken together, our study demonstrated that exosomes derived from miR‐375‐overexpressing hASCs promoted bone regeneration.
Collapse
Affiliation(s)
- Si Chen
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yiman Tang
- 4th Division, Peking University Hospital of Stomatology, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China
| | - Lingfei Jia
- National Clinical Research Center for Oral Diseases, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China
| |
Collapse
|
304
|
Lu J, Wang QY, Sheng JG. Exosomes in the Repair of Bone Defects: Next-Generation Therapeutic Tools for the Treatment of Nonunion. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1983131. [PMID: 31467871 PMCID: PMC6699293 DOI: 10.1155/2019/1983131] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022]
Abstract
Nonunion with bone defects, a common complication after long bone fracture, is a major challenge for orthopaedic surgeons worldwide because of the high incidence rate and difficulties in achieving successful treatment. Bone defects are the main complications of nonunion. The conventional biological treatments for nonunion with bone defects involve the use of autologous bone grafts or bone graft substitutes and cell-based therapy. Traditional nonunion treatments have always been associated with safety issues and various other complications. Bone grafts have limited autologous cancellous bone and there is a risk of infection. Additionally, problems with bone graft substitutes, including rejection and stimulation of bone formation, have been noted, and the health of the stem cell niche is a major consideration in cell-based therapy. In recent years, researchers have found that exosomes can be used to deliver functional RNA and mediate cell-to-cell communication, suggesting that exosomes may repair bone defects by regulating cells and cytokines involved in bone metabolism. In this review, we highlight the possible relationships between risk factors for nonunion and exosomes. Additionally, we discuss the roles of exosomes in bone metabolism and bone regeneration.
Collapse
Affiliation(s)
- Jian Lu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
- The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Qi-Yang Wang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jia-Gen Sheng
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
305
|
Branscome H, Paul S, Khatkar P, Kim Y, Barclay RA, Pinto DO, Yin D, Zhou W, Liotta LA, El-Hage N, Kashanchi F. Stem Cell Extracellular Vesicles and their Potential to Contribute to the Repair of Damaged CNS Cells. J Neuroimmune Pharmacol 2019; 15:520-537. [PMID: 31338754 DOI: 10.1007/s11481-019-09865-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/10/2019] [Indexed: 12/31/2022]
Abstract
Neurological diseases and disorders are leading causes of death and disability worldwide. Many of these pathologies are associated with high levels of neuroinflammation and irreparable tissue damage. As the global burden of these pathologies continues to rise there is a significant need for the development of novel therapeutics. Due to their multipotent properties, stem cells have broad applications for tissue repair; additionally, stem cells have been shown to possess both immunomodulatory and neuroprotective properties. It is now believed that paracrine factors, such as extracellular vesicles (EVs), play a critical role in the functionality associated with stem cells. The diverse biological cargo contained within EVs are proposed to mediate these effects and, to date, the reparative and regenerative effects of stem cell EVs have been demonstrated in a wide range of cell types. While a high potential for their therapeutic use exists, there is a gap of knowledge surrounding their characterization, mechanisms of action, and how they may regulate cells of the CNS. Here, we report the isolation, characterization, and functional assessment of EVs from two sources of human stem cells, mesenchymal stem cells and induced pluripotent stem cells. We demonstrate the ability of these EVs to enhance the processes of cellular migration and angiogenesis, which are critical for both normal cellular development as well as cellular repair. Furthermore, we investigate their reparative effects on damaged cells, specifically those with relevance to the central nervous system. Collectively, our data highlight the similarities and differences among these EV populations and support the view that stem cells EV can be used to repair or partially reverse cellular damage. Graphical Abstract Stem cell-derived Extracellular Vesicles (EVs) for repair of damaged cells. EVs isolated from human induced pluripotent stem cells and mesenchymal stem cells contribute to the partial reversal of phenotypes induced by different sources of cellular damage.
Collapse
Affiliation(s)
- Heather Branscome
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Discovery Hall Room 182, 10900 University Blvd, Manassas, VA, 20110, USA.,American Type Culture Collection (ATCC), Manassas, VA, USA
| | | | - Pooja Khatkar
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Discovery Hall Room 182, 10900 University Blvd, Manassas, VA, 20110, USA
| | - Yuriy Kim
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Discovery Hall Room 182, 10900 University Blvd, Manassas, VA, 20110, USA
| | - Robert A Barclay
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Discovery Hall Room 182, 10900 University Blvd, Manassas, VA, 20110, USA
| | - Daniel O Pinto
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Discovery Hall Room 182, 10900 University Blvd, Manassas, VA, 20110, USA
| | | | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Nazira El-Hage
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Discovery Hall Room 182, 10900 University Blvd, Manassas, VA, 20110, USA.
| |
Collapse
|
306
|
Saif B, Zhang W, Zhang X, Gu Q, Yang P. Sn-Triggered Two-Dimensional Fast Protein Assembly with Emergent Functions. ACS NANO 2019; 13:7736-7749. [PMID: 31244042 DOI: 10.1021/acsnano.9b01392] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The discovery of a general strategy for organizing functional proteins into stable nanostructures with the desired dimension, shape, and function is an important focus in developing protein-based self-assembled materials, but the scalable synthesis of such materials and transfer to other substrates remain great challenges. We herein tackle this issue by creating a two-dimensional metal-protein hybrid nanofilm that is flexible and cost-effective with reliable self-recovery, stability, and multifunctionality. As it differs from traditional metal ions, we discover the capability of Sn2+ to initiate fast amyloid-like protein assembly (occurring in seconds) by effectively reducing the disulfide bonds of native globular proteins. The Sn2+-initiated lysozyme aggregation at the air/water interface leads to droplet flattening, a result never before reported in a protein system, which finally affords a multifunctional 2D Sn-doped hybrid lysozyme nanofilm with an ultralarge area (e.g., 0.2 m2) within a few minutes. The hybrid film is distinctive in its ease of coating on versatile material surfaces with endurable chemical and mechanical stability, optical transparency, and diverse end uses in antimicrobial and photo-/electrocatalytic scaffolds. Our approach provides not only insights into the effect of tin ions on macroscopic self-assembly of proteins but also a controllable and scalable synthesis of a potential biomimic framework for biomedical and biocatalytic applications.
Collapse
Affiliation(s)
- Bassam Saif
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering , Shaanxi Normal University , Xi'an 710062 , P.R. China
| | - Wenxin Zhang
- School and Hospital of Stomatology , Tianjin Medical University , 12 Observatory Road , Tianjin 30070 , P.R. China
| | - Xu Zhang
- School and Hospital of Stomatology , Tianjin Medical University , 12 Observatory Road , Tianjin 30070 , P.R. China
| | - Quan Gu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering , Shaanxi Normal University , Xi'an 710062 , P.R. China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering , Shaanxi Normal University , Xi'an 710062 , P.R. China
| |
Collapse
|
307
|
Jafari D, Malih S, Eslami SS, Jafari R, Darzi L, Tarighi P, Samadikuchaksaraei A. The relationship between molecular content of mesenchymal stem cells derived exosomes and their potentials: Opening the way for exosomes based therapeutics. Biochimie 2019; 165:76-89. [PMID: 31302163 DOI: 10.1016/j.biochi.2019.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022]
Abstract
At least, more than half of our understanding of extracellular vesicles owes to the studies conducted over the past few years. When it became clear that the exosomes have various potentials in medicine, extensive research has focused on these potentials in a variety of areas including cancer, drug delivery and regenerative medicine. The growing understanding of molecular structure and functions of exosomes causes the vision to become brighter in the exosomes complexity, and our attitude toward these vesicles has undergone changes accordingly. Proteomic and transcriptomic studies on exosomes have highlighted their molecular diversity. In this review, we explicitly examine the exosomes composition, molecular structure and their therapeutic potentials in some diseases. Due to the very heterogeneous nature of exosomes, the process of their use as a therapeutic agent in the clinic has been challenged. We are still at the beginning of recognizing the molecular composition of exosomes and mechanisms that affect their physiology and biology. The growing trend of engineering of exosomes has shown a promising future to further utilize them in a different field. Molecular profiling of exosomes and their content for their related potentials in regenerative medicine should be done exactly for further defining a minimum content for specific therapeutic potentials.
Collapse
Affiliation(s)
- Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran; Student Research Committee, Iran University of Medical Sciences, Tehran, Iran.
| | - Sara Malih
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Seyed Sadegh Eslami
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasool Jafari
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Leila Darzi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Ali Samadikuchaksaraei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
308
|
Extracellular vesicles from human urine-derived stem cells prevent osteoporosis by transferring CTHRC1 and OPG. Bone Res 2019; 7:18. [PMID: 31263627 PMCID: PMC6594995 DOI: 10.1038/s41413-019-0056-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/19/2019] [Accepted: 05/04/2019] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis is a debilitating bone disease affecting millions of people. Here, we used human urine-derived stem cells (USCs), which were noninvasively harvested from unlimited and easily available urine, as a “factory” to obtain extracellular vesicles (USC-EVs) and demonstrated that the systemic injection of USC-EVs effectively alleviates bone loss and maintains bone strength in osteoporotic mice by enhancing osteoblastic bone formation and suppressing osteoclastic bone resorption. More importantly, the anti-osteoporotic properties of USC-EVs are not notably disrupted by the age, gender, or health condition (with or without osteoporosis) of the USC donor. Mechanistic studies determined that collagen triple-helix repeat containing 1 (CTHRC1) and osteoprotegerin (OPG) proteins are enriched in USC-EVs and required for USC-EV-induced pro-osteogenic and anti-osteoclastic effects. Our results suggest that autologous USC-EVs represent a promising novel therapeutic agent for osteoporosis by promoting osteogenesis and inhibiting osteoclastogenesis by transferring CTHRC1 and OPG.
Collapse
|
309
|
Zhao M, Dai W, Wang H, Xue C, Feng J, He Y, Wang P, Li S, Bai D, Shu R. Periodontal ligament fibroblasts regulate osteoblasts by exosome secretion induced by inflammatory stimuli. Arch Oral Biol 2019; 105:27-34. [PMID: 31247478 DOI: 10.1016/j.archoralbio.2019.06.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVES This study evaluated the role of human periodontal ligament fibroblasts (hPDLFs)-derived exosomes in periodontitis progression and discovered whether hPDLFs influence bone remodeling activity via exosome secretion. MATERIALS AND METHODS Exosomes were isolated and quantified from Porphyromonas gingivalis lipopolysaccharide (LPS)-treated primary hPDLFs and evaluated by western blotting, dynamic light scattering, and transmission electron microscopy. GW4869 was used to block exosome secretion in conditioned medium (CM). hPDLFs-derived CM, CM containing GW4869 (CM + GW4869) and exosomes were used to stimulate MG-63 cell lines. The expression levels of proinflammatory mediators, osteogenic genes, and osteoclastogenesis-related genes were measured by quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, western blotting, and alkaline phosphatase staining. RESULTS Exosome-enriched protein and total exosomal protein levels were higher in the LPS-treated group than in the vehicle controls. hPDLFs-derived exosomes were incorporated into MG-63 osteoblasts and slightly upregulated the expression of Interleukin-6 and tumor necrosis factor-alpha. CM and exosomes inhibited alkaline phosphatase, Collagen-I, Runt-related transcription factor 2, and Osteoprotegerin expression as well as ALP activity, and blocking exosome secretion by GW4869 eliminated the inhibitory effects. CONCLUSION These results indicate that LPS-pretreated hPDLFs induce inflammation and inhibit osteogenic activity of osteoblasts through secreting exosomes. This study provides a potential mechanism by which localized periodontal inflammation may influence bone remodeling by release exosomes.
Collapse
Affiliation(s)
- Mengyuan Zhao
- Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, PR China
| | - Wenyu Dai
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Hongzhe Wang
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Chaoran Xue
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Jie Feng
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Yiruo He
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Peiqi Wang
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Sijia Li
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Ding Bai
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China
| | - Rui Shu
- Department of Orthodontics, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, ChengDu, Sichuan, PR China.
| |
Collapse
|
310
|
The Impact of Plasma-Derived Microvesicles From a Femoral Fracture Animal Model on Osteoblast Function. Shock 2019; 53:78-87. [PMID: 31157718 DOI: 10.1097/shk.0000000000001336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The role of microvesicles (MVs) in transcellular signal transduction has been demonstrated in different studies. However, the potential modulatory role of MVs in fracture healing remains unclear. Therefore, we investigated the impact of plasma-derived MVs after a femoral fracture on cranial osteoblasts. A femoral fracture with intramedullary stabilization was induced in Sprague Dawley rats. The animals were killed 3 days (group A), 1 week (group B), or 2 weeks (group C) after trauma induction. Animals without trauma served as controls. Osteoblasts from the cranial bone of a neonatal Sprague Dawley rats were cultured and stimulated with either plasma-derived MVs or MV-free plasma of groups A to C. The effects of MVs on osteoblasts were analyzed by growth assay, metabolic assay, and quantitative real-time polymerase chain reaction for osteocalcin, RUNX2, and collagen 1A to test differentiation of osteoblasts. MVs were time-dependently incorporated in osteoblasts and localized mainly around the nucleus. MVs increased the viability of osteoblasts, particularly in the late phase after femoral fracture (group A, P = 0.0276; group B, P = 0.0295; group C, P = 0.0407). Late-phase differentiation of osteoblasts was not stimulated by MVs but was by MV-free plasma (osteocalcin, groups C vs. control, P = 0.0454). The levels of transforming growth factor β1 (P = 0.0320) and insulin-like growth factor 1 ( P = 0.0211) were significantly higher in plasma than in MVs. MVs seem to modulate the viability of osteoblasts but not to affect osteoblast differentiation. Further studies are warranted to determine the characteristics and interactions of MVs. Potentially, MVs might act as a diagnostic or therapeutic tool in cases of impairment of fracture healing.
Collapse
|
311
|
Stem cells in Osteoporosis: From Biology to New Therapeutic Approaches. Stem Cells Int 2019; 2019:1730978. [PMID: 31281368 PMCID: PMC6589256 DOI: 10.1155/2019/1730978] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis is a systemic disease that affects the skeleton, causing reduction of bone density and mass, resulting in destruction of bone microstructure and increased risk of bone fractures. Since osteoporosis is a disease affecting the elderly and the aging of the world's population is constantly increasing, it is expected that the incidence of osteoporosis and its financial burden on the insurance systems will increase continuously and there is a need for more understanding this condition in order to prevent and/or treat it. At present, available drug therapy for osteoporosis primarily targets the inhibition of bone resorption and agents that promote bone mineralization, designed to slow disease progression. Safe and predictable pharmaceutical means to increase bone formation have been elusive. Stem cell therapy of osteoporosis, as a therapeutic strategy, offers the promise of an increase in osteoblast differentiation and thus reversing the shift towards bone resorption in osteoporosis. This review is focused on the current views regarding the implication of the stem cells in the cellular and physiologic mechanisms of osteoporosis and discusses data obtained from stem cell-based therapies of osteoporosis in experimental animal models and the possibility of their future application in clinical trials.
Collapse
|
312
|
Hu Y, Xu R, Chen CY, Rao SS, Xia K, Huang J, Yin H, Wang ZX, Cao J, Liu ZZ, Tan YJ, Luo J, Xie H. Extracellular vesicles from human umbilical cord blood ameliorate bone loss in senile osteoporotic mice. Metabolism 2019; 95:93-101. [PMID: 30668962 DOI: 10.1016/j.metabol.2019.01.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Senile osteoporosis is one of the most common age-related diseases worldwide. Accumulating evidences have indicated that young blood can reverse age-related impairments. Extracellular vesicles (EVs) exert therapeutic effects in a variety of diseases by delivering bioactive molecules such as microRNAs (miRNAs). The aim of the study is to evaluate the therapeutic potential of EVs from human umbilical cord blood plasma (UCB-EVs) on senile osteoporosis and to preliminarily clarify the underlying mechanism. METHODS UCB-EVs were injected into the tail vein of aged (16 months old) male C57BL/6 mice. Microcomputed tomography was performed to evaluate bone mass and microarchitecture of mice. The osteogenic and osteoclastic activities were determined by quantitative real-time PCR (qRT-PCR), histological examination and western blot analysis. In vitro, qRT-PCR assay was undertaken to explore the enrichment levels of a number of miRNAs that have positive effects in reducing bone loss. The efficacy of UCB-EVs on osteoblastic differentiation of bone marrow mesenchymal stromal cells (BMSCs) and osteoclastogenesis of RAW264.7 cells were assessed by cytochemical staining. Gene and protein expression changes were detected by qRT-PCR and western blotting respectively. Meanwhile, the roles of the selected miRNA in the regulatory effects of UCB-EVs on BMSCs and RAW264.7 cells were evaluated by using specific miRNA inhibitor. RESULTS The intravenous injection of UCB-EVs for two months attenuated bone loss in old mice, as defined by increased trabecular and cortical bone mass, enhanced osteoblast formation and reduced osteoclast formation compared to the control mice. In vitro, UCB-EVs could promote the osteogenic differentiation of BMSCs and inhibit the osteoclastogenesis of RAW264.7 cells. Moreover, it was confirmed that miR-3960 was highly enriched in UCB-EVs and miR-3960 inhibitor reversed the stimulatory effect of UCB-EVs on osteoblastic differentiation of BMSCs. CONCLUSION Our findings indicate that UCB-EVs ameliorate age-related bone loss by stimulating bone formation and inhibiting bone resorption, and miR-3960 mediated the osteogenic effect of UCB-EVs on BMSCs. Thus, UCB-EVs may represent a promising agent for prevention of senile osteoporosis.
Collapse
Affiliation(s)
- Yin Hu
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Chun-Yuan Chen
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shan-Shan Rao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya Nursing School, Central South University, Changsha, Hunan 410013, China
| | - Kun Xia
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jie Huang
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hao Yin
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhen-Xing Wang
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jia Cao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zheng-Zhao Liu
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yi-Juan Tan
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Juan Luo
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hui Xie
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, Hunan 410008, China; Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
313
|
Kim S, Kim TM. Generation of mesenchymal stem-like cells for producing extracellular vesicles. World J Stem Cells 2019; 11:270-280. [PMID: 31171955 PMCID: PMC6545523 DOI: 10.4252/wjsc.v11.i5.270] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/02/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitor cells with therapeutic potential against autoimmune diseases, inflammation, ischemia, and metabolic disorders. Contrary to the previous conceptions, recent studies have revealed that the tissue repair and immunomodulatory functions of MSCs are largely attributed to their secretome, rather than their potential to differentiate into desired cell types. The composition of MSC secretome encompasses cytokines and growth factors, in addition to the cell-derived structures known as extracellular vesicles (EVs). EVs are membrane-enclosed nanoparticles that are capable of delivering biomolecules, and it is now believed that MSC-derived EVs are the major players that induce biological changes in the target tissues. Based on these EVs’ characteristics, the potential of EVs derived from MSC (MSC-EV) in terms of tissue regeneration and immune modulation has grown during the last decade. However, the use of MSCs for producing sufficient amount of EVs has not been satisfactory due to limitations in the cell growth and large variations among the donor cell types. In this regard, pluripotent stem cells (PSCs)-derived MSC-like cells, which can be robustly induced and expanded in vitro, have emerged as more accessible cell source that can overcome current limitations of using MSCs for EV production. In this review, we have highlighted the methods of generating MSC-like cells from PSCs and their therapeutic outcome in preclinical studies. Finally, we have also discussed future requirements for making this cell-free therapy clinically feasible.
Collapse
Affiliation(s)
- Soo Kim
- Brexogen Research Center, Brexogen Inc., Seoul, Songpa-gu 05718, South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do, Pyeongchang 25354, South Korea
| |
Collapse
|
314
|
Ukon Y, Makino T, Kodama J, Tsukazaki H, Tateiwa D, Yoshikawa H, Kaito T. Molecular-Based Treatment Strategies for Osteoporosis: A Literature Review. Int J Mol Sci 2019; 20:E2557. [PMID: 31137666 PMCID: PMC6567245 DOI: 10.3390/ijms20102557] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023] Open
Abstract
Osteoporosis is an unavoidable public health problem in an aging or aged society. Anti-resorptive agents (calcitonin, estrogen, and selective estrogen-receptor modulators, bisphosphonates, anti-receptor activator of nuclear factor κB ligand antibody along with calcium and vitamin D supplementations) and anabolic agents (parathyroid hormone and related peptide analogs, sclerostin inhibitors) have major roles in current treatment regimens and are used alone or in combination based on the pathological condition. Recent advancements in the molecular understanding of bone metabolism and in bioengineering will open the door to future treatment paradigms for osteoporosis, including antibody agents, stem cells, and gene therapies. This review provides an overview of the molecular mechanisms, clinical evidence, and potential adverse effects of drugs that are currently used or under development for the treatment of osteoporosis to aid clinicians in deciding how to select the best treatment option.
Collapse
Affiliation(s)
- Yuichiro Ukon
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahiro Makino
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Joe Kodama
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Hiroyuki Tsukazaki
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Daisuke Tateiwa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Hideki Yoshikawa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takashi Kaito
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
315
|
Wiklander OPB, Brennan MÁ, Lötvall J, Breakefield XO, El Andaloussi S. Advances in therapeutic applications of extracellular vesicles. Sci Transl Med 2019; 11:eaav8521. [PMID: 31092696 PMCID: PMC7104415 DOI: 10.1126/scitranslmed.aav8521] [Citation(s) in RCA: 629] [Impact Index Per Article: 104.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/26/2019] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are nanometer-sized, lipid membrane-enclosed vesicles secreted by most, if not all, cells and contain lipids, proteins, and various nucleic acid species of the source cell. EVs act as important mediators of intercellular communication that influence both physiological and pathological conditions. Given their ability to transfer bioactive components and surmount biological barriers, EVs are increasingly being explored as potential therapeutic agents. EVs can potentiate tissue regeneration, participate in immune modulation, and function as potential alternatives to stem cell therapy, and bioengineered EVs can act as delivery vehicles for therapeutic agents. Here, we cover recent approaches and advances of EV-based therapies.
Collapse
Affiliation(s)
- Oscar P B Wiklander
- Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
- Evox Therapeutics Limited, Medawar Centre, Robert Robinson Avenue, Oxford OX4 4HG, UK
| | - Meadhbh Á Brennan
- Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- INSERM UMR 1238, PhyOS, Faculty of Medicine, Université de Nantes, 44034 Nantes cedex 1, France
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Xandra O Breakefield
- Departments of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
- Evox Therapeutics Limited, Medawar Centre, Robert Robinson Avenue, Oxford OX4 4HG, UK
| |
Collapse
|
316
|
Hadjiargyrou M, Komatsu DE. The Therapeutic Potential of MicroRNAs as Orthobiologics for Skeletal Fractures. J Bone Miner Res 2019; 34:797-809. [PMID: 30866092 PMCID: PMC6536331 DOI: 10.1002/jbmr.3708] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/04/2019] [Accepted: 02/23/2019] [Indexed: 12/19/2022]
Abstract
The repair of a fractured bone is critical to the well-being of humans. Failure of the repair process to proceed normally can lead to complicated fractures, exemplified by either a delay in union or a complete nonunion. Both of these conditions lead to pain, the possibility of additional surgery, and impairment of life quality. Additionally, work productivity decreases, income is reduced, and treatment costs increase, resulting in financial hardship. Thus, developing effective treatments for these difficult fractures or even accelerating the normal physiological repair process is warranted. Accumulating evidence shows that microRNAs (miRNAs), small noncoding RNAs, can serve as key regulatory molecules of fracture repair. In this review, a brief description of the fracture repair process and miRNA biogenesis is presented, as well as a summary of our current knowledge of the involvement of miRNAs in physiological fracture repair, osteoporotic fractures, and bone defect healing. Further, miRNA polymorphisms associated with fractures, miRNA presence in exosomes, and miRNAs as potential therapeutic orthobiologics are also discussed. This is a timely review as several miRNA-based therapeutics have recently entered clinical trials for nonskeletal applications and thus it is incumbent upon bone researchers to explore whether miRNAs can become the next class of orthobiologics for the treatment of skeletal fractures.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY 11568-8000
| | - David E. Komatsu
- Department of Orthopaedics, Stony Brook University, Stony Brook, NY 11794-8181
| |
Collapse
|
317
|
Holliday LS, Truzman E, Zuo J, Han G, Torres-Medina R, Rody WJ. Extracellular vesicle identification in tooth movement models. Orthod Craniofac Res 2019; 22 Suppl 1:101-106. [PMID: 31074148 PMCID: PMC6512852 DOI: 10.1111/ocr.12287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs) are 30-150 nm in diameter vesicles released by cells that serve important intercellular regulatory functions. EVs include exosomes and microvesicles. Exosomes form in multivesicular bodies and are released extracellularly as the multivesicular bodies fuse with the plasma membrane. Microvesicles bud directly from the plasma membrane. Here, we examine methods that are available or emerging to detect and study EVs during orthodontic tooth movement (OTM). EV's involvement in regulating bone remodelling associated with OTM may be demonstrated by adding isolated EVs to an animal model to change the rate of tooth movement. Exosomes in multivesicular bodies might be detected by immunogold labelling of markers in sections from the tooth and jaw and detection by electron microscopy. Gingival crevicular fluid (GCF) is enriched in EVs. Detection and characterization of EVs released by osteoclasts during resorption have been described, and this information could be used to analyse EVs in OTM models. Regulatory EVs may be enriched in the GCF from teeth that are being moved or are undergoing root resorption. Emerging approaches, including nanoparticle tracking, ExoView and micro- and nanofluidics, show promise for studying EVs in the GCF. Techniques that amplify signal, including polymerase chain reaction (PCR), provide the sensitivity necessary to utilize EVs from GCF as biomarkers. Studies of the role of EVs in OTM will provide fresh insight that may identify means for enhancing OTM procedures. EVs in GCF may include biomarkers for bone remodelling during OTM, orthodontic-associated root resorption, and other dental pathologies.
Collapse
Affiliation(s)
- L Shannon Holliday
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
- Department of Anatomy & Cell Biology, University of Florida College of Medicine, Gainesville, Florida
| | - Estella Truzman
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
| | - Jian Zuo
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
| | - Guanghong Han
- Department of Oral Geriatrics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Rosemarie Torres-Medina
- Department of Orthodontics, University of Florida College of Dentistry, Gainesville, Florida
| | - Wellington J Rody
- Department of Orthodontics and Pediatric Dentistry, Stony Brook School of Dental Medicine, Stony Brook, New York
| |
Collapse
|
318
|
Humbert P, Brennan MÁ, Davison N, Rosset P, Trichet V, Blanchard F, Layrolle P. Immune Modulation by Transplanted Calcium Phosphate Biomaterials and Human Mesenchymal Stromal Cells in Bone Regeneration. Front Immunol 2019; 10:663. [PMID: 31001270 PMCID: PMC6455214 DOI: 10.3389/fimmu.2019.00663] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/11/2019] [Indexed: 12/22/2022] Open
Abstract
A wide variety of biomaterials have been developed as both stabilizing structures for the injured bone and inducers of bone neoformation. They differ in chemical composition, shape, porosity, and mechanical properties. The most extensively employed and studied subset of bioceramics are calcium phosphate materials (CaPs). These materials, when transplanted alongside mesenchymal stem cells (MSCs), lead to ectopic (intramuscular and subcutaneous) and orthotopic bone formation in preclinical studies, and effective fracture healing in clinical trials. Human MSC transplantation in pre-clinical and clinical trials reveals very low engraftment in spite of successful clinical outcomes and their therapeutic actions are thought to be primarily through paracrine mechanisms. The beneficial role of transplanted MSC could rely on their strong immunomodulatory effect since, even without long-term engraftment, they have the ability to alter both the innate and adaptive immune response which is critical to facilitate new bone formation. This study presents the current knowledge of the immune response to the implantation of CaP biomaterials alone or in combination with MSC. In particular the central role of monocyte-derived cells, both macrophages and osteoclasts, in MSC-CaP mediated bone formation is emphasized. Biomaterial properties, such as macroporosity and surface microstructure, dictate the host response, and the ultimate bone healing cascade. Understanding intercellular communications throughout the inflammation, its resolution and the bone regeneration phase, is crucial to improve the current therapeutic strategies or develop new approaches.
Collapse
Affiliation(s)
- Paul Humbert
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | - Meadhbh Á. Brennan
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Noel Davison
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- Instructure Labs, B.V., The Hague, Netherlands
| | - Philippe Rosset
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Centre Hospitalier Universitaire de Tours, Tours, France
| | - Valérie Trichet
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | | | - Pierre Layrolle
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| |
Collapse
|
319
|
Arafa MAA, Gouda ZA, El-Naseery NI, Abdel-Nour HM, Hanafy SM, Mohamed AF, Abo-Ouf AM. Bone Marrow-Derived Mesenchymal Stem Cells Ameliorate the Pancreatic Changes of Chemically Induced Hypothyroidism by Carbimazole in Male Rats. Cells Tissues Organs 2019; 206:144-156. [PMID: 30921798 DOI: 10.1159/000497297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/25/2019] [Indexed: 11/19/2022] Open
Abstract
We induced hypothyroidism (HT) in male rats through chronic oral administration of carbimazole and then tested whether an i.v. injection of rat bone marrow-derived mesenchymal stem cells (BM-MSCs) could ameliorate the HT-induced changes in pancreatic structure and function. The thyroid and pancreatic function tests, as well as total antioxidant capacity (TAC) and malondialdehyde (MDA) were estimated. The pancreatic structure was evaluated by hematoxylin and eosin (H&E) stain. Insulin protein and cleaved caspase-3 were detected immunohistochemically. The degree of apoptosis was assessed by TUNEL assay. The morphometric measurements were done by an image analyzer system and the obtained data were statistically analyzed. HT rats showed hyperglycemia associated with insulin deficiency, decreased TAC and increased MDA levels. H&E-stained sections showed that the pancreatic septa were infiltrated with acidophilic material. Some acini were vacuolated while others showed depleted acidophilia and dilated lumina. Spindle-shaped cells were accumulated within deformed islets in HT rats. The positive reaction with anti-cleaved caspase-3 was exclusively noted in the cytoplasm of islet cells with no immunostaining reaction in the acinar and ductal cells, whereas the positively stained nuclei with TUNEL were demonstrated in the islet and acinar cells. A significant increase in the apoptotic index % of both markers was detected. Injection of BM-MSCs in HT rats restored all biochemical indicators of disturbed pancreatic function to normal level and improved pancreatic structure, resulting in a clear septa and normal appearance of acini and islets. In conclusion, many of the significant structural and func tional pancreatic alterations detected in HT rats were ameliorated after the injection of BM-MSCs. These data demonstrate the ability of BM-MSCs to repair pancreatic disturbances. Further studies on humans are necessary to determine the potential clinical applications of BM-MSCs.
Collapse
Affiliation(s)
- Mona A A Arafa
- Department of Anatomy, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Zienab A Gouda
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt,
| | - Nesma I El-Naseery
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Hanim M Abdel-Nour
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Safaa M Hanafy
- Department of Anatomy, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Amany F Mohamed
- Department of Anatomy, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Amany M Abo-Ouf
- Department of Anatomy, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
320
|
Cao H, Yue Z, Gao H, Chen C, Cui K, Zhang K, Cheng Y, Shao G, Kong D, Li Z, Ding D, Wang Y. In Vivo Real-Time Imaging of Extracellular Vesicles in Liver Regeneration via Aggregation-Induced Emission Luminogens. ACS NANO 2019; 13:3522-3533. [PMID: 30844245 DOI: 10.1021/acsnano.8b09776] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Extracellular vesicles (EVs) attract much attention in liver pathology because they regulate cell-cell communication and many pathophysiological events by transferring their cargos. Monitoring and understanding the in vivo fate and therapeutic capacity of these EVs is critical for the development and optimization of EV-based diagnosis and therapy. Herein, we demonstrate the use of an aggregation-induced emission luminogen, DPA-SCP, for the real-time tracking of EVs derived from human placenta-derived mesenchymal stem cells (MSCs) and their therapeutic effects in a mouse acute liver injury (ALI) model. In vitro, DPA-SCP does not alter the inherent characteristics of MSC-derived EVs and shows extremely low toxicity. Moreover, DPA-SCP exhibited superior labeling efficiency and tracking capability to the most popular commercial EV trackers, PKH26 and DiI. In vivo, DPA-SCP precisely and quantitatively tracked the behaviors of EVs for 7 days in the mouse ALI model without influencing their regenerative capacity and therapeutic efficacy. The therapeutic effects of EVs may attribute to their ability for reducing inflammatory cell infiltration, enhancing cell survival and antiapoptotic effects. In conclusion, DPA-SCP with an AIE signature serves as a favorable and safe tracker for in vivo real-time imaging of EVs in liver regeneration.
Collapse
Affiliation(s)
- Hongmei Cao
- Nankai University School of Medicine , Tianjin 300071 , China
| | - Zhiwei Yue
- Nankai University School of Medicine , Tianjin 300071 , China
| | - Heqi Gao
- The Key Laboratory of Bioactive Materials, Ministry of Education , Nankai University, The College of Life Science , Tianjin 300071 , China
| | - Chao Chen
- The Key Laboratory of Bioactive Materials, Ministry of Education , Nankai University, The College of Life Science , Tianjin 300071 , China
| | - Kaige Cui
- Nankai University School of Medicine , Tianjin 300071 , China
| | - Kaiyue Zhang
- Nankai University School of Medicine , Tianjin 300071 , China
| | - Yuanqiu Cheng
- Nankai University School of Medicine , Tianjin 300071 , China
| | - Guoqiang Shao
- Department of Nuclear Medicine, Nanjing First Hospital , Nanjing Medical University , Nanjing 210006 , China
| | - Deling Kong
- The Key Laboratory of Bioactive Materials, Ministry of Education , Nankai University, The College of Life Science , Tianjin 300071 , China
| | - Zongjin Li
- Nankai University School of Medicine , Tianjin 300071 , China
| | - Dan Ding
- The Key Laboratory of Bioactive Materials, Ministry of Education , Nankai University, The College of Life Science , Tianjin 300071 , China
| | - Yuebing Wang
- Nankai University School of Medicine , Tianjin 300071 , China
| |
Collapse
|
321
|
Liu S, Mahairaki V, Bai H, Ding Z, Li J, Witwer KW, Cheng L. Highly Purified Human Extracellular Vesicles Produced by Stem Cells Alleviate Aging Cellular Phenotypes of Senescent Human Cells. Stem Cells 2019; 37:779-790. [PMID: 30811771 PMCID: PMC6767364 DOI: 10.1002/stem.2996] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/07/2019] [Accepted: 02/17/2019] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, mediate intercellular communications and exert various biological activities via delivering unique cargos of functional molecules such as RNAs and proteins to recipient cells. Previous studies showed that EVs produced and secreted by human mesenchymal stem cells (MSCs) can substitute intact MSCs for tissue repair and regeneration. In this study, we examined properties and functions of EVs from human induced pluripotent stem cells (iPSCs) that can be cultured infinitely under a chemically defined medium free of any exogenous EVs. We collected and purified EVs secreted by human iPSCs and MSCs. Purified EVs produced by both stem cell types have similar sizes (∼150 nm in diameter), but human iPSCs produced 16‐fold more EVs than MSCs. When highly purified iPSC‐EVs were applied in culture to senescent MSCs that have elevated reactive oxygen species (ROS), human iPSC‐EVs reduced cellular ROS levels and alleviated aging phenotypes of senescent MSCs. Our discovery reveals that EVs from human stem cells can alleviate cellular aging in culture, at least in part by delivering intracellular peroxiredoxin antioxidant enzymes. stem cells2019;37:779–790
Collapse
Affiliation(s)
- Senquan Liu
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Vasiliki Mahairaki
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Hao Bai
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zheng Ding
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jiaxin Li
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kenneth W. Witwer
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Linzhao Cheng
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
322
|
Zhu Y, Jia Y, Wang Y, Xu J, Chai Y. Impaired Bone Regenerative Effect of Exosomes Derived from Bone Marrow Mesenchymal Stem Cells in Type 1 Diabetes. Stem Cells Transl Med 2019; 8:593-605. [PMID: 30806487 PMCID: PMC6525563 DOI: 10.1002/sctm.18-0199] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022] Open
Abstract
Stem cell‐derived exosomes have exhibited promise for applications in tissue regeneration. However, one major problem for stem cell‐derived exosome therapies is identifying appropriate source cells. In the present study, we aimed to compare the bone regenerative effect of exosomes secreted by bone marrow mesenchymal stem cells (BMSCs) derived from type 1 diabetes rats (dBMSC‐exos) and exosomes secreted by BMSCs derived from normal rats (nBMSC‐exos). BMSCs were isolated from rats with streptozotocin‐induced diabetes and normal rats. dBMSC‐exos and nBMSC‐exos were isolated by an ultracentrifugation method and identified. The effects of dBMSC‐exos and nBMSC‐exos on the proliferation and migration of BMSCs and human umbilical vein endothelial cells (HUVECs) were investigated. The effects of exosomes on the osteogenic differentiation of BMSCs and the angiogenic activity of HUVECs were compared. Finally, a rat calvarial defect model was used to compare the effects of exosomes on bone regeneration and neovascularization in vivo. In vitro, dBMSC‐exos and nBMSC‐exos both enhanced the osteogenic differentiation of BMSCs and promoted the angiogenic activity of HUVECs, but nBMSC‐exos had a greater effect than dBMSC‐exos. Similarly, in vivo, both dBMSC‐exos and nBMSC‐exos promoted bone regeneration and neovascularization in rat calvarial defects, but the therapeutic effect of nBMSC‐exos was superior to that of dBMSC‐exos. The present study demonstrates for the first time that the bone regenerative effect of exosomes derived from BMSCs is impaired in type 1 diabetes, indicating that for patients with type 1 diabetes, the autologous transplantation of BMSC‐exos to promote bone regeneration may be inappropriate. stem cells translational medicine2019;8:593–605
Collapse
Affiliation(s)
- Yu Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Yachao Jia
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Yanmao Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| |
Collapse
|
323
|
Chamberlain CS, Clements AEB, Kink JA, Choi U, Baer GS, Halanski MA, Hematti P, Vanderby R. Extracellular Vesicle-Educated Macrophages Promote Early Achilles Tendon Healing. Stem Cells 2019; 37:652-662. [PMID: 30720911 PMCID: PMC6850358 DOI: 10.1002/stem.2988] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 12/14/2018] [Accepted: 01/21/2019] [Indexed: 12/20/2022]
Abstract
Tendon healing follows a complex series of coordinated events, which ultimately produces a mechanically inferior tissue more scar‐like than native tendon. More regenerative healing occurs when anti‐inflammatory M2 macrophages play a more dominant role. Mesenchymal stromal/stem cells (MSCs) are able to polarize macrophages to an M2 immunophenotype via paracrine mechanisms. We previously reported that coculture of CD14+ macrophages (MQs) with MSCs resulted in a unique M2‐like macrophage. More recently, we generated M2‐like macrophages using only extracellular vesicles (EVs) isolated from MSCs creating “EV‐educated macrophages” (also called exosome‐educated macrophages [EEMs]), thereby foregoing direct use of MSCs. For the current study, we hypothesized that cell therapy with EEMs would improve in vivo tendon healing by modulating tissue inflammation and endogenous macrophage immunophenotypes. We evaluated effects of EEMs using a mouse Achilles tendon rupture model and compared results to normal tendon healing (without any biologic intervention), MSCs, MQs, or EVs. We found that exogenous administration of EEMs directly into the wound promoted a healing response that was significantly more functional and more regenerative. Injured tendons treated with exogenous EEMs exhibited (a) improved mechanical properties, (b) reduced inflammation, and (c) earlier angiogenesis. Treatment with MSC‐derived EVs alone were less effective functionally but stimulated a biological response as evidenced by an increased number of endothelial cells and decreased M1/M2 ratio. Because of their regenerative and immunomodulatory effects, EEM treament could provide a novel strategy to promote wound healing in this and various other musculoskeletal injuries or pathologies where inflammation and inadequate healing is problematic. Stem Cells2019;37:652–662
Collapse
Affiliation(s)
- Connie S Chamberlain
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - Anna E B Clements
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - John A Kink
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Ugeun Choi
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| | - Geoffrey S Baer
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - Matthew A Halanski
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Ray Vanderby
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
324
|
Mancuso P, Raman S, Glynn A, Barry F, Murphy JM. Mesenchymal Stem Cell Therapy for Osteoarthritis: The Critical Role of the Cell Secretome. Front Bioeng Biotechnol 2019; 7:9. [PMID: 30761298 PMCID: PMC6361779 DOI: 10.3389/fbioe.2019.00009] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is an inflammatory condition still lacking effective treatments. Mesenchymal stem/stromal cells (MSCs) have been successfully employed in pre-clinical models aiming to resurface the degenerated cartilage. In early-phase clinical trials, intra-articular (IA) administration of MSCs leads to pain reduction and cartilage protection or healing. However, the consistent lack of engraftment indicates that the observed effect is delivered through a "hit-and-run" mechanism, by a temporal release of paracrine molecules. MSCs express a variety of chemokines and cytokines that aid in repair of degraded tissue, restoration of normal tissue metabolism and, most importantly, counteracting inflammation. Secretion of therapeutic factors is increased upon licensing by inflammatory signals or apoptosis, induced by the host immune system. Trophic effectors are released as soluble molecules or carried by extracellular vesicles (ECVs). This review provides an overview of the functions and mechanisms of MSC-secreted molecules found to be upregulated in models of OA, whether using in vitro or in vivo models.
Collapse
Affiliation(s)
- Patrizio Mancuso
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Swarna Raman
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Aoife Glynn
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Frank Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - J Mary Murphy
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biosciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
325
|
Zhu Y, Wang Y, Jia Y, Xu J, Chai Y. Catalpol promotes the osteogenic differentiation of bone marrow mesenchymal stem cells via the Wnt/β-catenin pathway. Stem Cell Res Ther 2019; 10:37. [PMID: 30670092 PMCID: PMC6341609 DOI: 10.1186/s13287-019-1143-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 01/03/2019] [Accepted: 01/09/2019] [Indexed: 12/22/2022] Open
Abstract
Background Rehmanniae Radix is a traditional herbal medicine in East Asia that has been widely used to treat patients with osteoporosis. However, the effect of catalpol, the primary active principle component of Rehmanniae Radix, on the function of bone marrow mesenchymal stem cells (BMSCs) and the underlying molecular mechanisms associated with its activity remain poorly understood. Methods The effect of catalpol on the proliferation of BMSCs was evaluated using a Cell Counting Kit-8 assay. Alkaline phosphatase (ALP) staining, ALP activity and Alizarin Red staining were performed to elucidate the effect of catalpol on the osteogenesis of BMSCs. qRT-PCR, Western blotting and immunofluorescence were performed to evaluate the expression of osteo-specific markers and the Wnt/β-catenin signalling-related genes and proteins. Moreover, a rat critical-sized calvarial defect model and a rat ovariectomy model were used to assess the effect of catalpol on bone regeneration in vivo. Results Catalpol significantly enhanced osteoblast-specific gene expression, alkaline phosphatase activity and calcium deposition in BMSCs in vitro. This phenomenon was accompanied by an upregulation of Wnt/β-catenin signalling. In addition, the enhanced osteogenesis due to catalpol treatment was partially reversed by a Wnt/β-catenin antagonist. Furthermore, catalpol increased the bone healing capacity of BMSCs in a rat critical-sized calvarial defect model and attenuated bone loss in a rat ovariectomy model. Conclusions These data suggest that catalpol enhances the osteogenic differentiation of BMSCs, partly via activation of the Wnt/β-catenin pathway. Catalpol may provide a new strategy for bone tissue engineering and can be a potential agent for the treatment of postmenopausal osteoporosis. Electronic supplementary material The online version of this article (10.1186/s13287-019-1143-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, People's Republic of China
| | - Yanmao Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, People's Republic of China
| | - Yachao Jia
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, People's Republic of China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, People's Republic of China.
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
326
|
Zuo R, Liu M, Wang Y, Li J, Wang W, Wu J, Sun C, Li B, Wang Z, Lan W, Zhang C, Shi C, Zhou Y. BM-MSC-derived exosomes alleviate radiation-induced bone loss by restoring the function of recipient BM-MSCs and activating Wnt/β-catenin signaling. Stem Cell Res Ther 2019; 10:30. [PMID: 30646958 PMCID: PMC6334443 DOI: 10.1186/s13287-018-1121-9] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023] Open
Abstract
Background Radiotherapy to cancer patients is inevitably accompanied by normal tissue injury, and the bone is one of the most commonly damaged tissues. Damage to bone marrow mesenchymal stem cells (BM-MSCs) induced by radiation is thought to be a major cause of radiation-induced bone loss. Exosomes exhibit great therapeutic potential in the treatment of osteoporosis, but whether exosomes are involved in radiation-induced bone loss has not been thoroughly elucidated to date. The main purpose of this study is to investigate the role of exosomes derived from BM-MSCs in restoring recipient BM-MSC function and alleviating radiation-induced bone loss. Methods BM-MSC-derived exosomes were intravenously injected to rats immediately after irradiation. After 28 days, the left tibiae were harvested for micro-CT and histomorphometric analysis. The effects of exosomes on antioxidant capacity, DNA damage repair, proliferation, and cell senescence of recipient BM-MSCs were determined. Osteogenic and adipogenic differentiation assays were used to detect the effects of exosomes on the differentiation potential of recipient BM-MSCs, and related genes were measured by qRT-PCR and Western blot analysis. β-Catenin expression was detected at histological and cytological levels. Results BM-MSC-derived exosomes can attenuate radiation-induced bone loss in a rat model that is similar to mesenchymal stem cell transplantation. Exosome-treated BM-MSCs exhibit reduced oxidative stress, accelerated DNA damage repair, and reduced proliferation inhibition and cell senescence-associate protein expression compared with BM-MSCs that exclusively received irradiation. Following irradiation, exosomes promote β-catenin expression in BM-MSCs and restore the balance between adipogenic and osteogenic differentiation. Conclusions Our findings indicate that BM-MSC-derived exosomes take effects by restoring the function of recipient BM-MSCs. Therefore, exosomes may represent a promising cell-free therapeutic approach for the treatment of radiation-induced bone loss. Electronic supplementary material The online version of this article (10.1186/s13287-018-1121-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Zuo
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Yanqiu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Wenkai Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Junlong Wu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chao Sun
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Bin Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University(Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Weiren Lan
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University(Third Military Medical University), Chongqing, 400038, People's Republic of China.
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China.
| |
Collapse
|
327
|
Huang T, Deng CX. Current Progresses of Exosomes as Cancer Diagnostic and Prognostic Biomarkers. Int J Biol Sci 2019; 15:1-11. [PMID: 30662342 PMCID: PMC6329932 DOI: 10.7150/ijbs.27796] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022] Open
Abstract
Cancer related exosomes are nano-size membrane vesicles that play important roles in tumor microenvironment. Emerging evidence indicates that exosomes can load unique cargoes, including proteins and nucleic acids that reflect the condition of tumor. Therefore, exosomes are being used as diagnostic and prognostic biomarkers for various cancers. In this review, we describe the current progresses of cancer related exosomes, including their biogenesis, molecular contents, biological functions, sources where they are derived from, and methods for their detection. We will also discuss the current exosomal biomarkers and the utilization of them for early diagnosis and prognostics in cancer.
Collapse
Affiliation(s)
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
328
|
Saberianpour S, Heidarzadeh M, Geranmayeh MH, Hosseinkhani H, Rahbarghazi R, Nouri M. Tissue engineering strategies for the induction of angiogenesis using biomaterials. J Biol Eng 2018; 12:36. [PMID: 30603044 PMCID: PMC6307144 DOI: 10.1186/s13036-018-0133-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is touted as a fundamental procedure in the regeneration and restoration of different tissues. The induction of de novo blood vessels seems to be vital to yield a successful cell transplantation rate loaded on various scaffolds. Scaffolds are natural or artificial substances that are considered as one of the means for delivering, aligning, maintaining cell connection in a favor of angiogenesis. In addition to the potential role of distinct scaffold type on vascularization, the application of some strategies such as genetic manipulation, and conjugation of pro-angiogenic factors could intensify angiogenesis potential. In the current review, we focused on the status of numerous scaffolds applicable in the field of vascular biology. Also, different strategies and priming approaches useful for the induction of pro-angiogenic signaling pathways were highlighted.
Collapse
Affiliation(s)
- Shirin Saberianpour
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 2Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Heidarzadeh
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
| | - Mohammad Hossein Geranmayeh
- 3Neuroscience Research Center, Imam Reza Medical Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 5Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- 2Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 5Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
329
|
Zhu Z, Zhang Y, Zhang Y, Zhang H, Liu W, Zhang N, Zhang X, Zhou G, Wu L, Hua K, Ding J. Exosomes derived from human umbilical cord mesenchymal stem cells accelerate growth of VK2 vaginal epithelial cells through MicroRNAsin vitro. Hum Reprod 2018; 34:248-260. [PMID: 30576496 DOI: 10.1093/humrep/dey344] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Zhongyi Zhu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yijing Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- Shanghai Ji Ai Genetics & IVF Institute, Shanghai, China
| | - Yiqun Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Hongdao Zhang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Wei Liu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Ning Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xiaodan Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Guannan Zhou
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Ligang Wu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Keqin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Jingxin Ding
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
330
|
Exosomes-the enigmatic regulators of bone homeostasis. Bone Res 2018; 6:36. [PMID: 30534458 PMCID: PMC6286319 DOI: 10.1038/s41413-018-0039-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a heterogeneous group of cell-derived membranous structures, which mediate crosstalk interaction between cells. Recent studies have revealed a close relationship between exosomes and bone homeostasis. It is suggested that bone cells can spontaneously secret exosomes containing proteins, lipids and nucleic acids, which then to regulate osteoclastogenesis and osteogenesis. However, the network of regulatory activities of exosomes in bone homeostasis as well as their therapeutic potential in bone injury remain largely unknown. This review will detail and discuss the characteristics of exosomes, the regulatory activities of exosomes in bone homeostasis as well as the clinical potential of exosomes in bone injury. Vesicles known as exosomes may prove to be valuable clinical tools once their function is clarified. Exosomes were discovered in the 1980s but not observed in bone tissue until 2003. Minghao Zheng of the University of Western Australia, together with colleagues elsewhere, has reviewed the biology of exosomes, their role in maintaining bones, and their potential clinical uses. Exosomes carry lipids, proteins, and nucleic acids between cells. They are released by every type of bone cell, with the role of each exosome determined by its specific contents. Exosome-mediated crosstalk is involved in regulating bone remodeling, and exosomes have also been implicated in myelomas. Recent work has shown that exosome treatment can improve fracture healing. The authors conclude that a better understanding of the role of exosomes in bone homeostasis will unlock their significant clinical potential.
Collapse
|
331
|
Aghebati‐Maleki L, Dolati S, Zandi R, Fotouhi A, Ahmadi M, Aghebati A, Nouri M, Kazem Shakouri S, Yousefi M. Prospect of mesenchymal stem cells in therapy of osteoporosis: A review. J Cell Physiol 2018; 234:8570-8578. [PMID: 30488448 DOI: 10.1002/jcp.27833] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Leili Aghebati‐Maleki
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| | - Sanam Dolati
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Reza Zandi
- Department of Orthopedic Surgery Faculty of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Ali Fotouhi
- Department of Orthopedic Surgery Faculty of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Majid Ahmadi
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Ali Aghebati
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Nouri
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Seyed Kazem Shakouri
- Physical Medicine and Rehabilitation Research Centre, Tabriz University of Medical Sciences Tabriz Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
332
|
Exosomal KLF3-AS1 from hMSCs promoted cartilage repair and chondrocyte proliferation in osteoarthritis. Biochem J 2018; 475:3629-3638. [PMID: 30341166 DOI: 10.1042/bcj20180675] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
The present study was designed to explore whether exosomal lncRNA-KLF3-AS1 derived from human mesenchymal stem cells (hMSCs) can serve as a positive treatment for osteoarthritis (OA). hMSCs and MSC-derived exosomes (MSC-exo) were prepared for morphological observation and identification by transmission electron microscopy and flow cytometry. IL-1β-induced OA chondrocytes and collagenase-induced rat model of OA were established for the further experiments. Lentivirus-mediated siRNA targeting KLF3-AS1 was transfected into MSCs for silencing KLF3-AS1. The real-time quantitative PCR and western blotting analysis were performed to examine the mRNA and protein levels of type II collagen alpha 1 (Col2a1), aggrecan, matrix metalloproteinase 13 and runt-related transcription factor 2. Cell proliferation, apoptosis and migration were evaluated by CCK-8 assay, flow cytometry and transwell assay. HE (hematoxylin and eosin) staining and immunohistochemistry were used for histopathological studies. MSC-exo ameliorated IL-1β-induced cartilage injury. Furthermore, lncRNA KLF3-AS1 was markedly enriched in MSC-exo, and exosomal KLF3-AS1 suppressed IL-1β-induced apoptosis of chondrocytes. Further in vivo investigation indicated that exosomal KLF3-AS1 promoted cartilage repair in a rat model of OA. Exosomal KLF3-AS1 promoted cartilage repair and chondrocyte proliferation in a rat model of OA, which might be an underlying therapeutic target for OA.
Collapse
|
333
|
Taheri B, Soleimani M, Fekri Aval S, Esmaeili E, Bazi Z, Zarghami N. Induced pluripotent stem cell-derived extracellular vesicles: A novel approach for cell-free regenerative medicine. J Cell Physiol 2018; 234:8455-8464. [PMID: 30478831 DOI: 10.1002/jcp.27775] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
In recent years, induced pluripotent stem cells (iPSCs) have been considered as a promising approach in the field of regenerative medicine. iPSCs can be generated from patients' somatic cells and possess the potential to differentiate, under proper conditions, into any cell type. However, the clinical application of iPS cells is restricted because of their tumorigenic potential. Recent studies have indicated that stem cells exert their therapeutic benefit via a paracrine mechanism, and extracellular vesicles have been demonstrated that play a critical role in this paracrine mechanism. Due to lower immunogenicity, easier management, and presenting no risk of tumor formation, in recent years, researchers turned attention to exosomes as potential alternatives to whole-cell therapy. Application of exosomes derived from iPSCs and their derived precursor provides a promising approach for personalized regenerative medicine. This study reviews the physiological functions of extracellular vesicles and discusses their potential therapeutic benefit in regenerative medicine.
Collapse
Affiliation(s)
- Behnaz Taheri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Technology Research Center, Tehran, Iran
| | | | - Sedigheh Fekri Aval
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Zahra Bazi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
334
|
Lin HT, Chen SK, Guo JW, Su IC, Huang CJ, Chien CC, Chang CJ. Dynamic expression of SMAD3 is critical in osteoblast differentiation of PDMCs. Int J Mol Med 2018; 43:1085-1093. [PMID: 30483761 DOI: 10.3892/ijmm.2018.4001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/19/2018] [Indexed: 11/06/2022] Open
Abstract
Human pluripotent stem cells have the potential assist in the identification of genes involved in mammalian development. The human placenta is considered a repository of stem cells, termed placenta‑derived multipotent cells (PDMCs), which are able to differentiate into cells with an osteoblastic phenotype. This plasticity of PDMCs maybe applied clinically to the understanding of osteogenesis and osteoporosis. In the presentstudy, osteoblasts were generated by culturing PDMCs in osteogenic medium. Reverse transcription quantitative polymerase chain reactionand the degree of osteoblast calcification were used to evaluate the efficacy of osteogenesis. The results suggestedthat the expression of mothers against decapentaplegic homolog 3 (SMAD3) increased in the initial stages of osteogenic differentiation but decreased in the later stages. However, osteogenesis was inhibitedwhen the PDMCs overexpressed SMAD3 throughout the differentiation period. In addition, the rate of osteogenic differentiation was decreased when SMAD3 signaling was impaired. In conclusion, SMAD3 serves an important role in osteoblast differentiation and bone formation in a time‑dependent manner. The data from the present study indicate that arapid increase in SMAD3 expression is crucial for osteogenesis and suggest a role for PDMCs in the treatment of patients with osteoporosis.
Collapse
Affiliation(s)
- Hsi-Ting Lin
- Department of Orthopedics, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Shao-Kuan Chen
- Department of Urology, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan, R.O.C
| | - Jiun-Wen Guo
- Ph.D. Program in Pharmaceutical Biotechnology, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - I-Chang Su
- Department of Neurosurgery, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan, R.O.C
| | - Chi-Jung Huang
- Ph.D. Program in Pharmaceutical Biotechnology, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Chih-Cheng Chien
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Chih-Ju Chang
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| |
Collapse
|
335
|
Liu Y, Lin L, Zou R, Wen C, Wang Z, Lin F. MSC-derived exosomes promote proliferation and inhibit apoptosis of chondrocytes via lncRNA-KLF3-AS1/miR-206/GIT1 axis in osteoarthritis. Cell Cycle 2018; 17:2411-2422. [PMID: 30324848 DOI: 10.1080/15384101.2018.1526603] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Exosomes secreted by human mesenchymal stem cells (hMSCs) have been shown to promote cartilage regeneration. This study aimed to explore whether exosomal lncRNA-KLF3-AS1 derived from hMSCs can promote chondrocyte proliferation via miR-206/GIT1 axis in osteoarthritis (OA). METHODS hMSCs and MSC-derived exosomes (MSC-exo) were prepared for morphological observation and identification by transmission electron microscopy (TEM) and flow cytometry. IL-1β-induced OA chondrocytes and collagenase-induced mouse OA model were established for the further experiments. Luciferase activity assay was performed to test whether miR-206 could bind to KLF3-AS1 or GIT1. Cell proliferation and apoptosis were evaluated by CCK-8 assay and flow cytometry, respectively. RESULTS MSC-Exos increased chondrogenic genes Col2a1 (type II collagen alpha 1) and aggrecan, decreased hondrocyte hypertrophy markers MMP-13 (matrix metalloproteinase-13) and Runx2 (runt-related transcription factor 2) in chondrocytes isolated from OA model mice. Furthermore, MSC-Exos attenuated IL-1β-induced chondrocyte proliferation inhibition and apoptosis induction. Moreover, MSCKLF3-AS1-Exos (exosomes derived from KLF3-AS1-overexpressing-MSCs) ameliorated IL-1β-induced chondrocyte injury. Results also demonstrated that KLF3-AS1 acted as a competitive endogenous RNA (ceRNA) by sponging miR-206 to facilitate GIT1 expression. In addition, miR-206 overexpression and GIT1 knockdown reversed MSCKLF3-AS1-Exos-mediated attenuation of chondrocyte injury. CONCLUSION Exosomal KLF3-AS1 derived from MSCs involved in MSC-Exos-mediated chondrocyte proliferation induction and chondrocyte apoptosis inhibition via miR-206/GIT1 axis. Abbreviation: G-protein-coupled receptor kinase interacting protein-1 (GIT1).
Collapse
Affiliation(s)
- Yubao Liu
- a Department of Orthopaedics , Luhe People's Hospital of Nanjing , Nanjing , China
| | - Lupan Lin
- a Department of Orthopaedics , Luhe People's Hospital of Nanjing , Nanjing , China
| | - Rui Zou
- a Department of Orthopaedics , Luhe People's Hospital of Nanjing , Nanjing , China
| | - Chuanyang Wen
- a Department of Orthopaedics , Luhe People's Hospital of Nanjing , Nanjing , China
| | - Zhen Wang
- a Department of Orthopaedics , Luhe People's Hospital of Nanjing , Nanjing , China
| | - Fuqing Lin
- a Department of Orthopaedics , Luhe People's Hospital of Nanjing , Nanjing , China
| |
Collapse
|
336
|
Lu M, Guo S, Hong F, Zhang Y, Yuan L, Ma C, Ma J. Pax2 is essential for proliferation and osteogenic differentiation of mouse mesenchymal stem cells via Runx2. Exp Cell Res 2018; 371:342-352. [DOI: 10.1016/j.yexcr.2018.08.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/19/2018] [Accepted: 08/21/2018] [Indexed: 01/09/2023]
|
337
|
Cengiz IF, Oliveira JM, Reis RL. Micro-CT - a digital 3D microstructural voyage into scaffolds: a systematic review of the reported methods and results. Biomater Res 2018; 22:26. [PMID: 30275969 PMCID: PMC6158835 DOI: 10.1186/s40824-018-0136-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/03/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Cell behavior is the key to tissue regeneration. Given the fact that most of the cells used in tissue engineering are anchorage-dependent, their behavior including adhesion, growth, migration, matrix synthesis, and differentiation is related to the design of the scaffolds. Thus, characterization of the scaffolds is highly required. Micro-computed tomography (micro-CT) provides a powerful platform to analyze, visualize, and explore any portion of interest in the scaffold in a 3D fashion without cutting or destroying it with the benefit of almost no sample preparation need. MAIN BODY This review highlights the relationship between the scaffold microstructure and cell behavior, and provides the basics of the micro-CT method. In this work, we also analyzed the original papers that were published in 2016 through a systematic search to address the need for specific improvements in the methods section of the papers including the amount of provided information from the obtained results. CONCLUSION Micro-CT offers a unique microstructural analysis of biomaterials, notwithstanding the associated challenges and limitations. Future studies that will include micro-CT characterization of scaffolds should report the important details of the method, and the derived quantitative and qualitative information can be maximized.
Collapse
Affiliation(s)
- Ibrahim Fatih Cengiz
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim Miguel Oliveira
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
| |
Collapse
|
338
|
Chen Z, Wang H, Xia Y, Yan F, Lu Y. Therapeutic Potential of Mesenchymal Cell-Derived miRNA-150-5p-Expressing Exosomes in Rheumatoid Arthritis Mediated by the Modulation of MMP14 and VEGF. THE JOURNAL OF IMMUNOLOGY 2018; 201:2472-2482. [PMID: 30224512 DOI: 10.4049/jimmunol.1800304] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/13/2018] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial tissue inflammation and joint destruction associated with the activation of angiogenesis. Exosomes, which play a role in cell-to-cell communication as carriers of genetic information, transfer microRNAs (miRNAs or miRs) between cells and have been studied as delivery vehicles for therapeutic molecules. The aim of the current study was to investigate the therapeutic effect of mesenchymal stem cell (MSC)-derived miR-150-5p exosomes on joint destruction in RA. The expression and secretion of miR-150-5p, matrix metalloproteinase (MMP) 14, and vascular endothelial growth factor (VEGF) in RA patients and fibroblast-like synoviocytes (FLS) were examined by quantitative RT-PCR, ELISA, and Western blotting. Immunohistochemistry was used to assess angiogenesis. MSCs were transfected with an miR-150-5p expression plasmid, and MSC-derived exosomes were harvested. The effect of MSC-derived miR-150-5p exosomes (Exo-150) on MMP14 and VEGF expression was examined. The effects of Exo-150 on cell migration and invasion in cytokine-stimulated FLS from RA patients were examined by HUVEC tube formation and transwell assays. The effect of Exo-150 in vivo was examined in a collagen-induced arthritis mouse model. Exo-150 decreased migration and invasion in RA FLS and downregulated tube formation in HUVECs by targeting MMP14 and VEGF. Injection of Exo-150 reduced hind paw thickness and the clinical arthritic scores in collagen-induced arthritis mice. Exo-150 reduced joint destruction by inhibiting synoviocyte hyperplasia and angiogenesis. Exosomes facilitate the direct intracellular transfer of miRNAs between cells and represent a potential therapeutic strategy for RA.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Hanqi Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Yang Xia
- Department of Physics, Oakland University, Rochester, MI 48309
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Yong Lu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| |
Collapse
|
339
|
Zhang K, Zhao X, Chen X, Wei Y, Du W, Wang Y, Liu L, Zhao W, Han Z, Kong D, Zhao Q, Guo Z, Han Z, Liu N, Ma F, Li Z. Enhanced Therapeutic Effects of Mesenchymal Stem Cell-Derived Exosomes with an Injectable Hydrogel for Hindlimb Ischemia Treatment. ACS APPLIED MATERIALS & INTERFACES 2018; 10:30081-30091. [PMID: 30118197 DOI: 10.1021/acsami.8b08449] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Mesenchymal stem cell (MSC)-derived exosomes have been recognized as new candidates for cell-free treatment of various diseases. However, maintaining the retention and stability of exosomes over time in vivo after transplantation is a major challenge in the clinical application of MSC-derived exosomes. Here, we investigated if human placenta-derived MSC-derived exosomes incorporated with chitosan hydrogel could boost the retention and stability of exosomes and further enhance their therapeutic effects. Our results demonstrated that chitosan hydrogel notably increased the stability of proteins and microRNAs in exosomes, as well as augmented the retention of exosomes in vivo as confirmed by Gaussia luciferase imaging. In addition, we assessed endothelium-protective and proangiogenesis abilities of hydrogel-incorporated exosomes in vitro. Meanwhile, we evaluated the therapeutic function of hydrogel-incorporated exosomes in a murine model of hindlimb ischemia. Our data demonstrated that chitosan hydrogel could enhance the retention and stability of exosomes and further augment the therapeutic effects for hindlimb ischemia as revealed by firefly luciferase imaging of angiogenesis. The strategy used in this study may facilitate the development of easy and effective approaches for assessing and enhancing the therapeutic effects of stem cell-derived exosomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Linan Liu
- Department of Pharmaceutical Sciences, Department of Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center & Edwards Lifesciences Center for Advanced Cardiovascular Technology, and Department of Biological Chemistry , University of California , Irvine 92697 , United States
| | - Weian Zhao
- Department of Pharmaceutical Sciences, Department of Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center & Edwards Lifesciences Center for Advanced Cardiovascular Technology, and Department of Biological Chemistry , University of California , Irvine 92697 , United States
| | - Zhibo Han
- Beijing Engineering Laboratory of Perinatal Stem Cells , Beijing Institute of Health and Stem Cells, Health & Biotech Co. , Beijing 100176 , China
- State Key Lab of Experimental Hematology , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300020 , China
| | | | | | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration , Xinxiang Medical University , Xinxiang 453003 , China
| | - Zhongchao Han
- Beijing Engineering Laboratory of Perinatal Stem Cells , Beijing Institute of Health and Stem Cells, Health & Biotech Co. , Beijing 100176 , China
| | | | - Fengxia Ma
- State Key Lab of Experimental Hematology , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300020 , China
| | | |
Collapse
|
340
|
Behera J, George AK, Voor MJ, Tyagi SC, Tyagi N. Hydrogen sulfide epigenetically mitigates bone loss through OPG/RANKL regulation during hyperhomocysteinemia in mice. Bone 2018; 114:90-108. [PMID: 29908298 PMCID: PMC6084464 DOI: 10.1016/j.bone.2018.06.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/06/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
Hydrogen sulfide (H2S) is a novel gasotransmitter produced endogenously in mammalian cells, which works by mediating diverse physiological functions. An imbalance in H2S metabolism is associated with defective bone homeostasis. However, it is unknown whether H2S plays any epigenetic role in bone loss induced by hyperhomocysteinemia (HHcy). We demonstrate that diet-induced HHcy, a mouse model of metabolite induced osteoporosis, alters homocysteine metabolism by decreasing plasma levels of H2S. Treatment with NaHS (H2S donor), normalizes the plasma level of H2S and further alleviates HHcy induced trabecular bone loss and mechanical strength. Mechanistic studies have shown that DNMT1 expression is higher in the HHcy condition. The data show that activated phospho-JNK binds to the DNMT1 promoter and causes epigenetic DNA hyper-methylation of the OPG gene. This leads to activation of RANKL expression and mediates osteoclastogenesis. However, administration of NaHS could prevent HHcy induced bone loss. Therefore, H2S could be used as a novel therapy for HHcy mediated bone loss.
Collapse
Affiliation(s)
- Jyotirmaya Behera
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40292, USA
| | - Akash K George
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40292, USA
| | - Michael J Voor
- Departments of Orthopaedic Surgery and Bioengineering, School of Medicine and Speed School of Engineering, University of Louisville, Louisville, KY 40292, USA
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40292, USA
| | - Neetu Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
341
|
Bjørge IM, Kim SY, Mano JF, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine - a new paradigm for tissue repair. Biomater Sci 2018; 6:60-78. [PMID: 29184934 DOI: 10.1039/c7bm00479f] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue regeneration by stem cells is driven by the paracrine activity of shedding vesicles and exosomes, which deliver specific cargoes to the recipient cells. Proteins, RNA, cytokines and subsequent gene expression, orchestrate the regeneration process by improving the microenvironment to promote cell survival, controlling inflammation, repairing injury and enhancing the healing process. The action of microRNA is widely accepted as an essential driver of the regenerative process through its impact on multiple downstream biological pathways, and its ability to regulate the host immune response. Here, we present an overview of the recent potential uses of exosomes for regenerative medicine and tissue engineering. We also highlight the differences in composition between shedding vesicles and exosomes that depend on the various types of stem cells from which they are derived. The conditions that affect the production of exosomes in different cell types are deliberated. This review also presents the current status of candidate exosomal microRNAs for potential therapeutic use in regenerative medicine, and in applications involving widely studied organs and tissues such as heart, lung, cartilage and bone.
Collapse
Affiliation(s)
- I M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | | | | | | | | |
Collapse
|
342
|
Liu W, Wang Y, Gong F, Rong Y, Luo Y, Tang P, Zhou Z, Zhou Z, Xu T, Jiang T, Yang S, Yin G, Chen J, Fan J, Cai W. Exosomes Derived from Bone Mesenchymal Stem Cells Repair Traumatic Spinal Cord Injury by Suppressing the Activation of A1 Neurotoxic Reactive Astrocytes. J Neurotrauma 2018; 36:469-484. [PMID: 29848167 DOI: 10.1089/neu.2018.5835] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell (MSC) transplantation is now considered as an effective treatment strategy for traumatic spinal cord injury (SCI). However, several key issues remain unresolved, including low survival rates, cell dedifferentiation, and tumor formation. Recent studies have demonstrated that the therapeutic effect of transplanted stem cells is primarily paracrine mediated. Exosomes are an important paracrine factor that can be used as a direct therapeutic agent. However, there are few reports on the application of exosomes derived from bone MSCs (BMSCs-Exos) in treating SCI. In this study, we demonstrated that BMSCs-Exos possessed robust proangiogenic properties, attenuated neuronal cells apoptosis, suppressed glial scar formation, attenuated lesion size, suppressed inflammation, promoted axonal regeneration, and eventually improved functional behavioral recovery effects after traumatic SCI. Briefly, lesion size was decreased by nearly 60%, neuronal apoptosis was attenuated by nearly 70%, glial scar formation was reduced by nearly 75%, average blood vessel density was increased by nearly 60%, and axonal regeneration was increased by almost 80% at day 28 after SCI in the BMSC-Exos group compared to the control group. Using a series of in vitro functional assays, we also confirmed that treatment with BSMCs-Exos significantly enhanced human umbilical vein endothelial cell proliferation, migration, and angiogenic tubule formation, attenuated neuronal cells apoptosis, and suppressed nitric oxide release in microglia. Moreover, our study demonstrated that administration of BMSCs-Exos suppressed inflammation efficiently after traumatic SCI and suppressed activation of A1 neurotoxic reactive astrocytes. In conclusion, our study suggested that the application of BMSCs-Exos may be a promising strategy for traumatic SCI.
Collapse
Affiliation(s)
- Wei Liu
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongxiang Wang
- 2 Department of Orthopaedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Fangyi Gong
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuluo Rong
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongjun Luo
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengyu Tang
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Zhou
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhimin Zhou
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xu
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Jiang
- 3 Department of Orthopaedics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Siting Yang
- 4 Department of Anesthesia, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoyong Yin
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Chen
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Fan
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weihua Cai
- 1 Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
343
|
Generation and Applications of Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells. Stem Cells Int 2018; 2018:9601623. [PMID: 30154868 PMCID: PMC6091255 DOI: 10.1155/2018/9601623] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/12/2018] [Accepted: 06/20/2018] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells with fibroblast-like morphology and isolated from the bone marrow via plastic adhesion. Their multipotency and immunoregulatory properties make MSCs possible therapeutic agents, and an increasing number of publications and clinical trials have highlighted their potential in regenerative medicine. However, the finite proliferative capacity of MSCs limits their scalability and global dissemination as a standardized therapeutic product. Furthermore, adult tissue provenance could constrain accessibility, impinge on cellular potency, and incur greater exposure to disease-causing pathogens based on the donor. These issues could be circumvented by the derivation of MSCs from pluripotent stem cells. In this paper, we review methods that induce and characterize MSCs derived from induced pluripotent stem cells (iPSCs) and introduce MSC applications to disease modeling, pathogenic mechanisms, and drug discovery. We also discuss the potential applications of MSCs in regenerative medicine including cell-based therapies and issues that should be overcome before iPSC-derived MSC therapy will be applied in the clinic.
Collapse
|
344
|
Exosomes: mediators of bone diseases, protection, and therapeutics potential. Oncoscience 2018; 5:181-195. [PMID: 30035185 PMCID: PMC6049320 DOI: 10.18632/oncoscience.421] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/06/2018] [Indexed: 12/15/2022] Open
Abstract
Bone remodeling is a continuous lifelong process in the repair of micro-damage to bone architecture and replacement of aging tissue in bone. A failure to such process leads to pathological destructive bone diseases such as osteoporosis, rheumatoid arthritis, and osteoarthritis. However, this active process is regulated by; osteoclasts, which are involved in the bone resorption process; osteoblasts, with involvement in the bone formation process and bone-derived endothelial cells, which promote angiogenesis. In the bone micro-environment, these cellular interactions are mediated by a complex interplay between cell types via direct interaction of cell secreted growth factors, such as cytokines. Recently, the discovery of exosomes (∼ 40–100 nm in size), has attracted more attention in the field of the bone remodeling process. Exosomes and microvesicles are derived from different types of bone cells such as mesenchymal stem cells, osteoblasts, osteoclasts and their precursors. They are also recognized to play pivotal roles in bone remodeling processes including osteogenesis, osteoclastogenesis, and angiogenesis. In this review, we especially emphasize the origin and biogenesis of exosomes and bone cell derived exosomes in the regulatory process of bone remodeling. Moreover, this review article also focuses on exosomal secreted proteins and microRNAs and their involvement in the regulation of bone remodeling.
Collapse
|
345
|
Fliefel R, Ehrenfeld M, Otto S. Induced pluripotent stem cells (iPSCs) as a new source of bone in reconstructive surgery: A systematic review and meta-analysis of preclinical studies. J Tissue Eng Regen Med 2018; 12:1780-1797. [DOI: 10.1002/term.2697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Riham Fliefel
- Experimental Surgery and Regenerative Medicine (ExperiMed), Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry; Alexandria University; Alexandria Egypt
| | - Michael Ehrenfeld
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| | - Sven Otto
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| |
Collapse
|
346
|
Rackov G, Garcia-Romero N, Esteban-Rubio S, Carrión-Navarro J, Belda-Iniesta C, Ayuso-Sacido A. Vesicle-Mediated Control of Cell Function: The Role of Extracellular Matrix and Microenvironment. Front Physiol 2018; 9:651. [PMID: 29922170 PMCID: PMC5996101 DOI: 10.3389/fphys.2018.00651] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) — including exosomes, microvesicles and apoptotic bodies — have received much scientific attention last decade as mediators of a newly discovered cell-to-cell communication system, acting at short and long distances. EVs carry biologically active molecules, thus providing signals that influence a spectrum of functions in recipient cells during various physiological and pathological processes. Recent findings point to EVs as very attractive immunomodulatory therapeutic agents, vehicles for drug delivery and diagnostic and prognostic biomarkers in liquid biopsies. In addition, EVs interact with and regulate the synthesis of extracellular matrix (ECM) components, which is crucial for organ development and wound healing, as well as bone and cardiovascular calcification. EVs carrying matrix metalloproteinases (MMPs) are involved in ECM remodeling, thus modifying tumor microenvironment and contributing to premetastatic niche formation and angiogenesis. Here we review the role of EVs in control of cell function, with emphasis on their interaction with ECM and microenvironment in health and disease.
Collapse
Affiliation(s)
| | | | - Susana Esteban-Rubio
- Fundación de Investigación HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad CEU San Pablo, Madrid, Spain
| | | | | | - Angel Ayuso-Sacido
- IMDEA Nanoscience Institute, Madrid, Spain.,Fundación de Investigación HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad CEU San Pablo, Madrid, Spain
| |
Collapse
|
347
|
Jing H, He X, Zheng J. Exosomes and regenerative medicine: state of the art and perspectives. Transl Res 2018; 196:1-16. [PMID: 29432720 DOI: 10.1016/j.trsl.2018.01.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 12/19/2022]
Abstract
Exosomes have attracted the attention of the scientific community in recent years due to their widespread distribution, their possible functions as biomarkers of disease, and their great potential to be applied as therapeutic agents. Exosomes carry proteins and nucleic acids that can facilitate their uptake by distant target cells through endocytosis, such that exosomes could be targeted to a specific cell or cells to enhance or interfere with specific biological processes. This review will mainly focus on their roles in tissue repair and regenerative processes. Exosomal engineering and their potential applications in tissue regeneration are also reviewed here as an outlook for future research.
Collapse
Affiliation(s)
- Hui Jing
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomin He
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jinghao Zheng
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
348
|
Deng H, Sun C, Sun Y, Li H, Yang L, Wu D, Gao Q, Jiang X. Lipid, Protein, and MicroRNA Composition Within Mesenchymal Stem Cell-Derived Exosomes. Cell Reprogram 2018; 20:178-186. [PMID: 29782191 DOI: 10.1089/cell.2017.0047] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) were regarded as one of the most promising type of seed cells in tissue engineering due to its easy accessibility and multipotent feature of being able to differentiate into adipocyte, osteoblast, cardiomyocytes, and neurons. For years, MSCs have been applied in treating cardiovascular disease, reconstructing kidney injury, and remodeling immune system with remarkable achievements. Basic researches revealed that its clinic effects are not only due to their pluripotent ability but also through their paracrine function that they synthesize and secrete a broad spectrum of growth factors and cytokines. Recent studies show that exosomes is the main paracrine executor of MSCs. The lipid bilayer of exosome maintains its stability and integrity and keeps biological potency of biological substance within it. MSC-derived exosomes were shown to be successful in treating many diseases, including tumor and cardiovascular diseases. However, the exact composition of MSC-derived exosomes is not known yet. In this review, we will discuss the lipid, protein, and microRNA contents within MSC-derived exosomes based on current studies to guide further research and clinical applications of MSC-derived exosomes.
Collapse
Affiliation(s)
- Hao Deng
- 1 First Teaching Hospital of Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Chun Sun
- 2 School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Yingxin Sun
- 2 School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Huhu Li
- 2 School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Lin Yang
- 2 School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Danbin Wu
- 2 School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Qing Gao
- 2 School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin, China
| | - Xijuan Jiang
- 2 School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin, China
| |
Collapse
|
349
|
Abstract
Large bone defects remain a tremendous clinical challenge. There is growing evidence in support of treatment strategies that direct defect repair through an endochondral route, involving a cartilage intermediate. While culture-expanded stem/progenitor cells are being evaluated for this purpose, these cells would compete with endogenous repair cells for limited oxygen and nutrients within ischaemic defects. Alternatively, it may be possible to employ extracellular vesicles (EVs) secreted by culture-expanded cells for overcoming key bottlenecks to endochondral repair, such as defect vascularization, chondrogenesis, and osseous remodelling. While mesenchymal stromal/stem cells are a promising source of therapeutic EVs, other donor cells should also be considered. The efficacy of an EV-based therapeutic will likely depend on the design of companion scaffolds for controlled delivery to specific target cells. Ultimately, the knowledge gained from studies of EVs could one day inform the long-term development of synthetic, engineered nanovesicles. In the meantime, EVs harnessed from in vitro cell culture have near-term promise for use in bone regenerative medicine. This narrative review presents a rationale for using EVs to improve the repair of large bone defects, highlights promising cell sources and likely therapeutic targets for directing repair through an endochondral pathway, and discusses current barriers to clinical translation. Cite this article: E. Ferreira, R. M. Porter. Harnessing extracellular vesicles to direct endochondral repair of large bone defects. Bone Joint Res 2018;7:263-273. DOI: 10.1302/2046-3758.74.BJR-2018-0006.
Collapse
Affiliation(s)
- E. Ferreira
- Departments of Internal Medicine and Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - R. M. Porter
- Departments of Internal Medicine and Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
350
|
Li W, Liu Y, Zhang P, Tang Y, Zhou M, Jiang W, Zhang X, Wu G, Zhou Y. Tissue-Engineered Bone Immobilized with Human Adipose Stem Cells-Derived Exosomes Promotes Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2018; 10:5240-5254. [PMID: 29359912 DOI: 10.1021/acsami.7b17620] [Citation(s) in RCA: 287] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Exosomes, nanoscale extracellular vesicles functioning as cell-to-cell communicators, are an emerging promising therapeutic in the field of bone tissue engineering. Here, we report the construction and evaluation of a novel cell-free tissue-engineered bone that successfully accelerated the restoration of critical-sized mouse calvarial defects through combining exosomes derived from human adipose-derived stem cells (hASCs) with poly(lactic-co-glycolic acid) (PLGA) scaffolds. The exosomes were immobilized on the polydopamine-coating PLGA (PLGA/pDA) scaffolds under mild chemical conditions. Specifically, we investigated the effects of hASC-derived exosomes on the osteogenic, proliferation, and migration capabilities of human bone marrow-derived mesenchymal stem cells in vitro and optimized their osteoinductive effects through osteogenic induction. Furthermore, an in vitro assay showed exosomes could release from PLGA/pDA scaffold slowly and consistently and in vivo results showed this cell-free system enhanced bone regeneration significantly, at least partially through its osteoinductive effects and capacities of promoting mesenchymal stem cells migration and homing in the newly formed bone tissue. Therefore, overall results demonstrated that our novel cell-free system comprised of hASC-derived exosomes and PLGA/pDA scaffold provides a new therapeutic paradigm for bone tissue engineering and showed promising potential in repairing bone defects.
Collapse
Affiliation(s)
| | | | | | | | - Miao Zhou
- Key laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University , Guangzhou 510140, China
| | | | | | - Gang Wu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit Amsterdam , Amsterdam 1081 LA, The Netherlands
| | | |
Collapse
|