1
|
Deng L, Liu Y, Wu Q, Lai S, Yang Q, Mu Y, Dong M. Exosomes to exosome-functionalized scaffolds: a novel approach to stimulate bone regeneration. Stem Cell Res Ther 2024; 15:407. [PMID: 39521993 PMCID: PMC11550564 DOI: 10.1186/s13287-024-04024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Bone regeneration is a complex biological process that relies on the orchestrated interplay of various cellular and molecular events. Bone tissue engineering is currently the most promising method for treating bone regeneration. However, the immunogenicity, stable and cell quantity of seed cells limited their application. Recently, exosomes, which are small extracellular vesicles released by cells, have been found to effectively address these problems and better induce bone regeneration. Meanwhile, a growing line of research has shown the cargos of exosomes may provide effective therapeutic and biomarker tools for bone repair, including miRNA, lncRNA, and proteins. Moreover, engineered scaffolds loaded with exosomes can offer a cell-free bone repair strategy, addressing immunogenicity concerns and providing a more stable functional performance. Herein, we provide a comprehensive summary of the role played by scaffolds loaded with exosomes in bone regeneration, drawing on a systematic analysis of relevant literature available on PubMed, Scopus, and Google Scholar database.
Collapse
Affiliation(s)
- Li Deng
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yang Liu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Qian Wu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Shuang Lai
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qiu Yang
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yandong Mu
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China.
| |
Collapse
|
2
|
Suciu TS, Feștilă D, Berindan-Neagoe I, Nutu A, Armencea G, Aghiorghiesei AI, Vulcan T, Băciuț M. Circular RNA-Mediated Regulation of Oral Tissue-Derived Stem Cell Differentiation: Implications for Oral Medicine and Orthodontic Applications. Stem Cell Rev Rep 2024; 20:656-671. [PMID: 38279054 PMCID: PMC10984898 DOI: 10.1007/s12015-024-10683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2024] [Indexed: 01/28/2024]
Abstract
Circular RNAs (circRNAs) are a novel class of endogenous non-coding RNAs (ncRNAs) which unlike linear RNAs, have a covalently closed continuous loop structure. circRNAs are found abundantly in human cells and their biology is complex. They feature unique expression to different types of cells, tissues, and developmental stages. To the present, the functional roles of circular RNAs are not fully understood. They reportedly act as microRNA (miRNA) sponges, therefore having key regulatory functions in diverse physiological and pathological processes. As for dentistry field, lines of evidence indicate that circRNAs play vital roles in the odontogenic and osteogenic differentiation of dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs). Abnormal expression of circRNAs have been found in other areas of pathology frequently reflected also in the oral environment, such as inflammation or bone and soft tissue loss. Therefore, circRNAs could be of significant importance in various fields in dentistry, especially in bone and soft tissue engineering and regeneration. Understanding the molecular mechanisms occurring during the regulation of oral biological and tissue remodeling processes could augment the discovery of novel diagnostic biomarkers and therapeutic strategies that will improve orthodontic and other oral therapeutic protocols.
Collapse
Affiliation(s)
- Tudor-Sergiu Suciu
- Department of Orthodontics and Dentofacial Orthopedics, Iuliu Hațieganu University of Medicine and Pharmacy, 400083, Cluj-Napoca, Romania
| | - Dana Feștilă
- Department of Orthodontics and Dentofacial Orthopedics, Iuliu Hațieganu University of Medicine and Pharmacy, 400083, Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | - Andreea Nutu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | - Gabriel Armencea
- Department of Maxillofacial Surgery and Implantology, Iuliu Hațieganu University of Medicine and Pharmacy, 400029, Cluj-Napoca, Romania
| | - Alexandra Iulia Aghiorghiesei
- Department of Prosthodontics and Dental Materials, Iuliu Hațieganu University of Medicine and Pharmacy, 400006, Cluj-Napoca, Romania
| | - Talida Vulcan
- Department of Dermatology, Iuliu Hațieganu University of Medicine and Pharmacy, 400006, Cluj-Napoca, Romania
| | - Mihaela Băciuț
- Department of Maxillofacial Surgery and Implantology, Iuliu Hațieganu University of Medicine and Pharmacy, 400029, Cluj-Napoca, Romania
| |
Collapse
|
3
|
Huang X, Deng Y, Xiao J, Wang H, Yang Q, Cao Z. Genetically engineered M2-like macrophage-derived exosomes for P. gingivalis-suppressed cementum regeneration: From mechanism to therapy. Bioact Mater 2024; 32:473-487. [PMID: 37965240 PMCID: PMC10640966 DOI: 10.1016/j.bioactmat.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/21/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
Cementum, a thin layer of mineralized tissue covering tooth root surface, is recognized as the golden standard in periodontal regeneration. However, current efforts mainly focus on alveolar bone regeneration rather than cementum regeneration, and rarely take Porphyromonas gingivalis (Pg), the keystone pathogen responsible for periodontal tissue destruction, into consideration. Though M2 macrophage-derived exosomes (M2-EXO) show promise in tissue regeneration, the exosome-producing M2 macrophages are induced by exogenous cytokines with transitory and unstable effects, restricting the regeneration potential of M2-EXO. Here, exosomes derived from genetically engineered M2-like macrophages are constructed by silencing of casein kinase 2 interacting protein-1 (Ckip-1), a versatile player involved in various biological processes. Ckip-1 silencing is proved to be an effective gene regulation strategy to obtain permanent M2-like macrophages with mineralization-promoting effect. Further, exosomes derived from Ckip-1-silenced macrophages (sh-Ckip-1-EXO) rescue Pg-suppressed cementoblast mineralization and cementogenesis. Mechanismly, sh-Ckip-1-EXO delivers Let-7f-5p targeting and silencing Ckip-1, a negative regulator also for cementum formation and cementoblast mineralization. More deeply, downregulation of Ckip-1 in cementoblasts by exosomal Let-7f-5p activates PGC-1α-dependent mitochondrial biogenesis. In all, this study provides a new strategy of genetically engineered M2-like macrophage-derived exosomes for cementum regeneration under Pg-dominated inflammation.
Collapse
Affiliation(s)
- Xin Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yifei Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junhong Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huiyi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qiudong Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Wang X, Gong W, Li R, Li L, Wang J. Preparation of genetically or chemically engineered exosomes and their therapeutic effects in bone regeneration and anti-inflammation. Front Bioeng Biotechnol 2024; 12:1329388. [PMID: 38314353 PMCID: PMC10834677 DOI: 10.3389/fbioe.2024.1329388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/11/2024] [Indexed: 02/06/2024] Open
Abstract
The treatment of bone or cartilage damage and inflammation-related diseases has been a long-standing research hotspot. Traditional treatments such as surgery and cell therapy have only displayed limited efficacy because they can't avoid potential deterioration and ensure cell activity. Recently, exosomes have become a favorable tool for various tissue reconstruction due to their abundant content of proteins, lipids, DNA, RNA and other substances, which can promote bone regeneration through osteogenesis, angiogenesis and inflammation modulation. Besides, exosomes are also promising delivery systems because of stability in the bloodstream, immune stealth capacity, intrinsic cell-targeting property and outstanding intracellular communication. Despite having great potential in therapeutic delivery, exosomes still show some limitations in clinical studies, such as inefficient targeting ability, low yield and unsatisfactory therapeutic effects. In order to overcome the shortcomings, increasing studies have prepared genetically or chemically engineered exosomes to improve their properties. This review focuses on different methods of preparing genetically or chemically engineered exosomes and the therapeutic effects of engineering exosomes in bone regeneration and anti-inflammation, thereby providing some references for future applications of engineering exosomes.
Collapse
Affiliation(s)
- Xinyue Wang
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Weitao Gong
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Rongrong Li
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Lin Li
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Jing Wang
- School of Stomatology, Lanzhou University, Lanzhou, China
- Clinical Research Center for Oral Diseases, Lanzhou, China
| |
Collapse
|
5
|
Ren S, Lin Y, Liu W, Yang L, Zhao M. MSC-Exos: Important active factor of bone regeneration. Front Bioeng Biotechnol 2023; 11:1136453. [PMID: 36814713 PMCID: PMC9939647 DOI: 10.3389/fbioe.2023.1136453] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
Bone defect and repair is a common but difficult problem in restorative and reconstructive surgery. Bone tissue defects of different sizes caused by different reasons bring functional limitations and cosmetic deformities to patients. Mesenchymal stem cells (MSC), a major hotspot in the field of regeneration in recent years, have been widely used in various studies on bone tissue regeneration. Numerous studies have shown that the bone regenerative effects of MSC can be achieved through exosome-delivered messages. Although its osteogenic mechanism is still unclear, it is clear that MSC-Exos can directly or indirectly support the action of bone regeneration. It can act directly on various cells associated with osteogenesis, or by carrying substances that affect cellular activators or the local internal environment in target cells, or it can achieve activation of the osteogenic framework by binding to materials. Therefore, this review aims to summarize the types and content of effective contents of MSC-Exos in bone regeneration, as well as recent advances in the currently commonly used methods to enable the binding of MSC-Exos to the framework and to conclude that MSC-Exos is effective in promoting osteogenesis.
Collapse
Affiliation(s)
- Sihang Ren
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China,Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China,NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University), Liaoning Research Institute of Family Planning (The Affiliated Reproductive Hospital of China Medical University), Shenyang, China
| | - Yuyang Lin
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Wenyue Liu
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Liqun Yang
- NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University), Liaoning Research Institute of Family Planning (The Affiliated Reproductive Hospital of China Medical University), Shenyang, China,Department of Biomaterials, Shengjing Hospital of China Medical University, Shenyang, China,*Correspondence: Liqun Yang, ; Muxin Zhao,
| | - Muxin Zhao
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China,*Correspondence: Liqun Yang, ; Muxin Zhao,
| |
Collapse
|
6
|
Ma S, Zhang Y, Li S, Li A, Li Y, Pei D. Engineering exosomes for bone defect repair. Front Bioeng Biotechnol 2022; 10:1091360. [PMID: 36568296 PMCID: PMC9768454 DOI: 10.3389/fbioe.2022.1091360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Currently, bone defect repair is still an intractable clinical problem. Numerous treatments have been performed, but their clinical results are unsatisfactory. As a key element of cell-free therapy, exosome is becoming a promising tool of bone regeneration in recent decades, because of its promoting osteogenesis and osteogenic differentiation function in vivo and in vitro. However, low yield, weak activity, inefficient targeting ability, and unpredictable side effects of natural exosomes have limited the clinical application. To overcome the weakness, various approaches have been applied to produce engineering exosomes by regulating their production and function at present. In this review, we will focus on the engineering exosomes for bone defect repair. By summarizing the exosomal cargos affecting osteogenesis, the strategies of engineering exosomes and properties of exosome-integrated biomaterials, this work will provide novel insights into exploring advanced engineering exosome-based cell-free therapy for bone defect repair.
Collapse
Affiliation(s)
| | | | | | | | - Ye Li
- *Correspondence: Ye Li, ; Dandan Pei,
| | | |
Collapse
|
7
|
Kim SI, Ha JY, Choi SY, Hong SH, Lee HJ. Use of Bacterial Extracellular Vesicles for Gene Delivery to Host Cells. Biomolecules 2022; 12:biom12091171. [PMID: 36139009 PMCID: PMC9496234 DOI: 10.3390/biom12091171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Extracellular vesicles (EVs), which are nanosized membranous particles secreted from both prokaryotic and eukaryotic cells, can deliver various biological molecules, such as nucleic acids, proteins, and lipids, into recipient cells. However, contrary to what is known about eukaryotic EVs, whether bacterial EVs (bEVs) can be used as transporters for bioactive molecules is becoming a hot area of research. In this study, we electroporated enhanced green fluorescent protein (EGFP) genes and precursor microRNA of Cel-miR-39 (pre-Cel-miR-39) from isolated bEVs of Escherichia coli and Lactobacillus reuteri. The EGFP plasmid, synthetic EGFP RNA, and pre-Cel-miR-39 were successfully delivered into the murine microglial BV2 cells via bEVs. PCR and confocal microscopy analysis confirmed the transfer of the EGFP plasmid and RNA. The bEV-delivered exogenous pre-Cel-miR-39 was further processed into the mature form of Cel-miR-39; its incorporation into Ago2—a major component of the RNA-induced silencing complex—was assessed using RNA-immunoprecipitation–PCR. Taken together, bEVs can be used as vehicles to deliver genetic materials and for novel biotechnological applications, such as gene transfer and mRNA vaccines.
Collapse
Affiliation(s)
- Su-Im Kim
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Jae Yeong Ha
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
- Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu 41940, Korea
| | - Song-Yi Choi
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
- Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu 41940, Korea
| | - Su-Hyung Hong
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Heon-Jin Lee
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
- Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu 41940, Korea
- Correspondence:
| |
Collapse
|
8
|
Fu W, Li T, Chen H, Zhu S, Zhou C. Research Progress in Exosome-Based Nanoscale Drug Carriers in Tumor Therapies. Front Oncol 2022; 12:919279. [PMID: 35800056 PMCID: PMC9253528 DOI: 10.3389/fonc.2022.919279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
Current antitumor treatment methods have several reported limitations, including multidrug resistance and serious adverse reactions. Targeted drug delivery systems are effective alternatives that can help healthcare providers overcome these limitations. Exosomes can serve as a natural nanoscale drug delivery system, with the advantages of high biocompatibility, low immunogenicity, and efficient tumor targetability. In this paper, we discuss the biological characteristics of exosomes, summarize the drug-carrying mechanisms of exosome-based drug delivery systems, and examine the potential role and applicability of exosomes in clinical tumor treatment approaches. This review can be used as a guideline for the future development of exosome-based delivery systems in clinical precision tumor treatment.
Collapse
Affiliation(s)
- Wei Fu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Chen
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Changkai Zhou, ; Shu Zhu, ; Hongbo Chen,
| | - Shu Zhu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Changkai Zhou, ; Shu Zhu, ; Hongbo Chen,
| | - Changkai Zhou
- Department of Burn and Plastic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People’s Hospital, Nantong, China
- *Correspondence: Changkai Zhou, ; Shu Zhu, ; Hongbo Chen,
| |
Collapse
|
9
|
Bin-Bin Z, Da-Wa ZX, Chao L, Lan-Tao Z, Tao W, Chuan L, Chao-Zheng L, De-Chun L, Chang F, Shu-Qing W, Zu-Nan D, Xian-Wei P, Zhang ZX, Ke-Wen L. M2 macrophagy-derived exosomal miRNA-26a-5p induces osteogenic differentiation of bone mesenchymal stem cells. J Orthop Surg Res 2022; 17:137. [PMID: 35246197 PMCID: PMC8895825 DOI: 10.1186/s13018-022-03029-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/18/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells have always been a heated research topic in bone tissue regeneration and repair because of their self-renewal and multi-differentiation potential. A large number of studies have been focused on finding the inducing factors that will promote the osteogenic differentiation of bone marrow mesenchymal stem cells. Previous studies have shown that macrophage exosomes or miRNA-26a-5p can make it work, but the function of this kind of substance on cell osteogenic differentiation has not been public. METHODS M2 macrophages are obtained from IL-4 polarized bone marrow-derived macrophages. Exosomes were isolated from the supernatant of M2 macrophages and identified via transmission electron microscopy (TEM), western blotting, and DLS. Chondrogenic differentiation potential was detected by Alcian blue staining. Oil red O staining was used to detect the potential for lipogenic differentiation. And MTT would detect the proliferative capacity of cells. Western blot was performed to detect differential expression of osteogenic differentiation-related proteins. RESULTS The results showed that M2 macrophage exosomes will promote bone differentiation and at the same time inhibit lipid differentiation. In addition, M2 macrophage-derived exosomes have the function of promoting the expression of SOX and Aggrecan suppressing the level of MMP13. The exosome inhibitor GW4689 suppresses miRNA-26a-5p in M2 macrophage exosomes, and the treated exosomes do not play an important role in promoting bone differentiation. Moreover, miRNA-26a-5p can enable to promote bone differentiation and inhibit lipid differentiation. miRNA-26a-5p can promote the expression of ALP (alkaline phosphatase), RUNX-2 (Runt-related transcription factor 2), OPN(osteopontin), and Col-2(collagen type II). Therefore, it is speculated that exosomal miRNA-26a-5p is indispensable in osteogenic differentiation. CONCLUSIONS The present study indicated that M2 macrophage exosomes carrying miRNA-26a-5p can induce osteogenic differentiation of bone marrow-derived stem cells to inhibit lipogenic differentiation, and miRNA-26a-5p will also promote the expression of osteogenic differentiation-related proteins ALP, RUNX-2, OPN, and Col-2.
Collapse
Affiliation(s)
- Zhang Bin-Bin
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zha Xi Da-Wa
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Li Chao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zhang Lan-Tao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Wu Tao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Lu Chuan
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Liu Chao-Zheng
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Li De-Chun
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Feng Chang
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Wei Shu-Qing
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Dong Zu-Nan
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Pei Xian-Wei
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zhi-Xia Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Out-Patient, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Li Ke-Wen
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China.
| |
Collapse
|
10
|
Lee KS, Lee J, Kim HK, Yeom SH, Woo CH, Jung YJ, Yun YE, Park SY, Han J, Kim E, Sul JH, Jung JM, Park JH, Choi JS, Cho YW, Jo D. Extracellular vesicles from adipose tissue-derived stem cells alleviate osteoporosis through osteoprotegerin and miR-21-5p. J Extracell Vesicles 2021; 10:e12152. [PMID: 34596354 PMCID: PMC8485335 DOI: 10.1002/jev2.12152] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 09/06/2021] [Accepted: 09/15/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoporosis is one of the most common skeletal disorders caused by the imbalance between bone formation and resorption, resulting in quantitative loss of bone tissue. Since stem cell-derived extracellular vesicles (EVs) are growing attention as novel cell-free therapeutics that have advantages over parental stem cells, the therapeutic effects of EVs from adipose tissue-derived stem cells (ASC-EVs) on osteoporosis pathogenesis were investigated. ASC-EVs were isolated by a multi-filtration system based on the tangential flow filtration (TFF) system and characterized using transmission electron microscopy, dynamic light scattering, zeta potential, flow cytometry, cytokine arrays, and enzyme-linked immunosorbent assay. EVs are rich in growth factors and cytokines related to bone metabolism and mesenchymal stem cell (MSC) migration. In particular, osteoprotegerin (OPG), a natural inhibitor of receptor activator of nuclear factor-κB ligand (RANKL), was highly enriched in ASC-EVs. We found that the intravenous administration of ASC-EVs attenuated bone loss in osteoporosis mice. Also, ASC-EVs significantly inhibited osteoclast differentiation of macrophages and promoted the migration of bone marrow-derived MSCs (BM-MSCs). However, OPG-depleted ASC-EVs did not show anti-osteoclastogenesis effects, demonstrating that OPG is critical for the therapeutic effects of ASC-EVs. Additionally, small RNA sequencing data were analysed to identify miRNA candidates related to anti-osteoporosis effects. miR-21-5p in ASC-EVs inhibited osteoclast differentiation through Acvr2a down-regulation. Also, let-7b-5p in ASC-EVs significantly reduced the expression of genes related to osteoclastogenesis. Finally, ASC-EVs reached the bone tissue after they were injected intravenously, and they remained longer. OPG, miR-21-5p, and let-7b-5p in ASC-EVs inhibit osteoclast differentiation and reduce gene expression related to bone resorption, suggesting that ASC-EVs are highly promising as cell-free therapeutic agents for osteoporosis treatment.
Collapse
Affiliation(s)
- Kyoung Soo Lee
- Department of Materials Science and Chemical EngineeringHanyang University ERICAAnsanKorea
- Exostemtech, Inc.AnsanKorea
| | - Jeongmi Lee
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | | | | | | | | | - Ye Eun Yun
- Department of Materials Science and Chemical EngineeringHanyang University ERICAAnsanKorea
| | | | - Jihoon Han
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Eunae Kim
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Jae Hoon Sul
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Jae Min Jung
- School of Chemical EngineeringCollege of EngineeringSungkyunkwan UniversitySuwonKorea
| | - Jae Hyung Park
- Exostemtech, Inc.AnsanKorea
- School of Chemical EngineeringCollege of EngineeringSungkyunkwan UniversitySuwonKorea
- Biomedical Institute for ConvergenceSungkyunkwan UniversitySuwonKorea
- Department of Health Science and TechnologySAIHSTSungkyunkwan UniversitySeoulKorea
| | | | - Yong Woo Cho
- Department of Materials Science and Chemical EngineeringHanyang University ERICAAnsanKorea
- Exostemtech, Inc.AnsanKorea
| | - Dong‐Gyu Jo
- Exostemtech, Inc.AnsanKorea
- School of PharmacySungkyunkwan UniversitySuwonKorea
- Biomedical Institute for ConvergenceSungkyunkwan UniversitySuwonKorea
- Department of Health Science and TechnologySAIHSTSungkyunkwan UniversitySeoulKorea
| |
Collapse
|
11
|
Rankin-Turner S, Vader P, O'Driscoll L, Giebel B, Heaney LM, Davies OG. A call for the standardised reporting of factors affecting the exogenous loading of extracellular vesicles with therapeutic cargos. Adv Drug Deliv Rev 2021; 173:479-491. [PMID: 33862168 PMCID: PMC8191593 DOI: 10.1016/j.addr.2021.04.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are complex nanoparticles required for the intercellular transfer of diverse biological cargoes. Unlike synthetic nanoparticles, EVs may provide a natural platform for the enhanced targeting and functional transfer of therapeutics across complex and often impenetrable biological boundaries (e.g. the blood-brain barrier or the matrix of densely organised tumours). Consequently, there is considerable interest in utilising EVs as advanced drug delivery systems for the treatment of a range of challenging pathologies. Within the past decade, efforts have focused on providing standard minimal requirements for conducting basic EV research. However, no standard reporting framework has been established governing the therapeutic loading of EVs for drug delivery applications. The purpose of this review is to critically evaluate progress in the field, providing an initial set of guidelines that can be applied as a benchmark to enhance reproducibility and increase the likelihood of translational outcomes.
Collapse
Affiliation(s)
- Stephanie Rankin-Turner
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK
| | - Pieter Vader
- CDL Research, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; Department of Experimental Cardiology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraβe 179, 45147 Essen, Germany
| | - Liam M Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK
| | - Owen G Davies
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK.
| |
Collapse
|
12
|
Exosome-Derived Noncoding RNAs as a Promising Treatment of Bone Regeneration. Stem Cells Int 2021; 2021:6696894. [PMID: 33542737 PMCID: PMC7843188 DOI: 10.1155/2021/6696894] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 02/05/2023] Open
Abstract
The reconstruction of large bone defects remains a crucial challenge in orthopedic surgery. The current treatments including autologous and allogenic bone grafting and bioactive materials have their respective drawbacks. While mesenchymal stem cell (MSC) therapy may address these limitations, growing researches have demonstrated that the effectiveness of MSC therapy depends on paracrine factors, particularly exosomes. This aroused great focus on the exosome-based cell-free therapy in the treatment of bone defects. Exosomes can transfer various cargoes, and noncoding RNAs are the most widely studied cargo through which exosomes exert their ability of osteoinduction. Here, we review the research status of the exosome-derived noncoding RNAs in bone regeneration, the potential application of exosomes, and the existing challenges.
Collapse
|
13
|
Xie L, Chen J, Ren X, Zhang M, Thuaksuban N, Nuntanaranont T, Guan Z. Alteration of circRNA and lncRNA expression profile in exosomes derived from periodontal ligament stem cells undergoing osteogenic differentiation. Arch Oral Biol 2020; 121:104984. [PMID: 33217605 DOI: 10.1016/j.archoralbio.2020.104984] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE This study investigated circRNA and lncRNA expression profile in exosomes derived from periodontal ligament stem cell (PDLSC) before and after its osteogenic differentiation. DESIGN Exosomes derived from PDLSCs before (EX0) and after osteogenic induction for 5 (EX5) and 7 (EX7) days were harvested and exosomal circRNAs and lncRNAs were analyzed by RNA sequencing. Certain RNAs showing significantly altered expression were selected for qRT-PCR verification. The circRNA-miRNA-mRNA network and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. RESULTS All groups of exosomes showed typical characteristics under nanoparticle tracking analysis, flow cytometry assay and transmission electron microscopy. 69-557 circRNAs and 2907-11581 lncRNAs were found in EX0, EX5 and EX7, which were broadly distributed across the 24 pairs of human chromosomes. Compared with EX0, 3 circRNAs and 2 lncRNAs were up-regulated and 39 circRNAs and 5 lncRNAs down-regulated consistently through out of EX5 and EX7, p < 0.05. qRT-PCR confirmed certain those consistently expressed RNAs, such as circ lysophosphatidic acid receptor 1 (LPAR1). KEGG analysis showed that those consistent expressed RNAs closely related to TGF-beta pathway, MAPK pathway, mTOR pathway and FoxO signaling pathways regulating pluripotency of stem cells. CONCLUSIONS Exosomal circRNAs and lncRNAs had significant expression changes during the early phase of osteogenic differentiation of PDLSCs. Further study would be taken for understanding the roles of exosomal circRNAs and lncRNAs playing in osteogenic differentiation of PDLSCs.
Collapse
Affiliation(s)
- Liangkun Xie
- Department of Oral Implantology, the Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Jianzhong Chen
- Department of Oral Anatomy and Physiology, School of Stomatology, Kunming Medical University, Kunming, China
| | - Xiaobin Ren
- Department of Periodontology, the Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Mingzhu Zhang
- Department of Periodontology, the Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Nattawut Thuaksuban
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Thongchai Nuntanaranont
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hatyai, Songkhla, Thailand.
| | - Zheng Guan
- Biomedical Research Center, the Affiliated Calmette Hospital of Kunming Medical University (the First Hospital of Kunming), Kunming, China.
| |
Collapse
|
14
|
Exosomal circLPAR1 Promoted Osteogenic Differentiation of Homotypic Dental Pulp Stem Cells by Competitively Binding to hsa-miR-31. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6319395. [PMID: 33062690 PMCID: PMC7539105 DOI: 10.1155/2020/6319395] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/29/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022]
Abstract
Human dental pulp stem cells (DPSCs) hold great promise in bone regeneration. However, the exact mechanism of osteogenic differentiation of DPSCs remains unknown, especially the role of exosomes played in. The DPSCs were cultured and received osteogenic induction; then, exosomes from osteogenic-induced DPSCs (OI-DPSC-Ex) at different time intervals were isolated and sequenced for circular RNA (circRNA) expression profiles. Gradually, increased circular lysophosphatidic acid receptor 1 (circLPAR1) expression was found in the OI-DPSC-Ex coincidentally with the degree of osteogenic differentiation. Meanwhile, results from osteogenic differentiation examinations showed that the OI-DPSC-Ex had osteogenic effect on the recipient homotypic DPSCs. To investigate the mechanism of exosomal circLPAR1 on osteogenic differentiation, we verified that circLPAR1 could competently bind to hsa-miR-31, by eliminating the inhibitory effect of hsa-miR-31 on osteogenesis, therefore promoting osteogenic differentiation of the recipient homotypic DPSCs. Our study showed that exosomal circRNA played an important role in osteogenic differentiation of DPSCs and provided a novel way of utilization of exosomes for the treatment of bone deficiencies.
Collapse
|
15
|
Al-Sowayan B, Alammari F, Alshareeda A. Preparing the Bone Tissue Regeneration Ground by Exosomes: From Diagnosis to Therapy. Molecules 2020; 25:E4205. [PMID: 32937850 PMCID: PMC7570455 DOI: 10.3390/molecules25184205] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
Bone tissue engineering employs acellular scaffolds or scaffolds, along with cells and growth factors, to provide the mechanical support needed, as well as serve as a delivery vehicle for bioactive molecules to the injury sites. As tissue engineering continues to evolve, it has integrated two emerging fields: stem cells and nanotechnology. A paracrine factor that is found to be responsible for the major regenerative effect in stem cell transplantation is an extracellular vesicle called an 'exosome'. Recent advances in nanotechnology have allowed the 'exosome' to be distinguished from other extracellular vesicles and be polymerized into a well-defined concept. Scientists are now investigating exosome uses in clinical applications. For bone-related diseases, exosomes are being explored as biomarkers for different bone pathologies. They are also being explored as a therapeutic agent where progenitor cell-derived exosomes are used to regenerate damaged bone tissue. In addition, exosomes are being tested as immune modulators for bone tissue inflammation, and finally as a delivery vehicle for therapeutic agents. This review discusses recently published literature on the clinical utilization of exosomes in bone-related applications and the correlated advantages. A particular focus will be placed on the potential utilization of regenerative cell-derived exosomes as a natural biomaterial for tissue regeneration.
Collapse
Affiliation(s)
- Batla Al-Sowayan
- Stem Cells and Regenerative Medicine Unit, Cell Therapy & Cancer Research Department, King Abdullah International Medical Research Center/King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| | | | - Alaa Alshareeda
- Stem Cells and Regenerative Medicine Unit, Cell Therapy & Cancer Research Department, King Abdullah International Medical Research Center/King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh 11481, Saudi Arabia;
| |
Collapse
|
16
|
Yoshiko Y, Minamizaki T. Emerging roles of microRNAs as extracellular vesicle cargo secreted from osteoblasts. J Oral Biosci 2020; 62:228-234. [PMID: 32535286 DOI: 10.1016/j.job.2020.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Extracellular vesicles (EVs) have come into the spotlight as messengers, delivering cargo for cell-cell communication. Concomitantly, increasing attention has been focused on microRNAs (miRNAs) as EV cargo. Besides their well-known role in extracellular matrix mineralization, whether matrix vesicles (MVs) - which are in a broad sense a class of EV - also deliver miRNAs to regulate the function of recipient cells remains unclear. HIGHLIGHT We recently found that MVs budding from osteoblasts contain many miRNAs that can be transferred to the bone matrix. Of these, miR-125b was released into the bone marrow microenvironment during bone resorption, where it targeted the transcriptional repressor Prdm1 in osteoclast precursors, resulting in increased expression of anti-osteoclastogenic factors and suppression of osteoclastogenesis, thereby increasing bone mass in mice. CONCLUSION Beyond their well-established action in bone mineralization, MVs play a role in the transport of miRNAs from osteoblasts into the bone matrix. Similar to the miR-125b axis in osteoclastogenesis, it seems likely that other miRNAs that accumulate in bone via MV transport may also act as mediators of cell-cell communication in the skeletal system.
Collapse
Affiliation(s)
- Yuji Yoshiko
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| | - Tomoko Minamizaki
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| |
Collapse
|
17
|
Abstract
IMPACT STATEMENT The number of commensal bacteria in the body surpasses the number of actual human cells. Thus, various interactions between microbes and human cells constitute an inevitable phenomenon. Recent evidence has led to bacterial extracellular RNAs (exRNAs) being proposed as good candidates for microbe-host inter-kingdom communication tools as they can modulate the expression of host genes. However, research findings on the relevance of interactions between extracellular RNA and human diseases are still in their infancy. Nevertheless, substantial data suggest that microbial exRNAs are implicated in various human diseases both at local and distant sites. By exploring various scenarios for the involvement of microbial exRNAs in human diseases, we may better understand the role of exRNAs as "communication signals" for diseases and thereby develop novel therapeutic strategies by using them and their carrier extracellular vesicles.
Collapse
Affiliation(s)
- Heon-Jin Lee
- Department of Microbiology and Immunology, Kyungpook National University School of Dentistry, Daegu 41940, Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu 41940, Korea
| |
Collapse
|
18
|
Han EC, Choi SY, Lee Y, Park JW, Hong SH, Lee HJ. Extracellular RNAs in periodontopathogenic outer membrane vesicles promote TNF-α production in human macrophages and cross the blood-brain barrier in mice. FASEB J 2019; 33:13412-13422. [PMID: 31545910 DOI: 10.1096/fj.201901575r] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Among the main bacteria implicated in the pathology of periodontal disease, Aggregatibacter actinomycetemcomitans (Aa) is well known for causing loss of periodontal attachment and systemic disease. Recent studies have suggested that secreted extracellular RNAs (exRNAs) from several bacteria may be important in periodontitis, although their role is unclear. Emerging evidence indicates that exRNAs circulate in nanosized bilayered and membranous extracellular vesicles (EVs) known as outer membrane vesicles (OMVs) in gram-negative bacteria. In this study, we analyzed the small RNA expression profiles in activated human macrophage-like cells (U937) infected with OMVs from Aa and investigated whether these cells can harbor exRNAs of bacterial origin that have been loaded into the host RNA-induced silencing complex, thus regulating host target transcripts. Our results provide evidence for the cytoplasmic delivery and activity of microbial EV-derived small exRNAs in host gene regulation. The production of TNF-α was promoted by exRNAs via the TLR-8 and NF-κB signaling pathways. Numerous studies have linked periodontal disease to neuroinflammatory diseases but without elucidating specific mechanisms for the connection. We show here that intracardiac injection of Aa OMVs in mice showed successful delivery to the brain after crossing the blood-brain barrier, the exRNA cargos increasing expression of TNF-α in the mouse brain. The current study indicates that host gene regulation by microRNAs originating from OMVs of the periodontal pathogen Aa is a novel mechanism for host gene regulation and that the transfer of OMV exRNAs to the brain may cause neuroinflammatory diseases like Alzheimer's.-Han, E.-C., Choi, S.-Y., Lee, Y., Park, J.-W., Hong, S.-H., Lee, H.-J. Extracellular RNAs in periodontopathogenic outer membrane vesicles promote TNF-α production in human macrophages and cross the blood-brain barrier in mice.
Collapse
Affiliation(s)
- Eun-Chong Han
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Song-Yi Choi
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Youngkyun Lee
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Jin-Woo Park
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Su-Hyung Hong
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Heon-Jin Lee
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
19
|
Hu X, Zhong Y, Kong Y, Chen Y, Feng J, Zheng J. Lineage-specific exosomes promote the odontogenic differentiation of human dental pulp stem cells (DPSCs) through TGFβ1/smads signaling pathway via transfer of microRNAs. Stem Cell Res Ther 2019; 10:170. [PMID: 31196201 PMCID: PMC6567518 DOI: 10.1186/s13287-019-1278-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022] Open
Abstract
Background Exosomes derived from dental pulp stem cells (DPSCs) can be used as biomimetic tools to induce odontogenic differentiation of stem cells, but the regulatory mechanisms and functions of exosome-encapsulated microRNAs are still unknown. The present study aimed to clarify the role of microRNAs contained in the exosomes derived from human DPSCs and their potential signaling cascade in odontogenic differentiation. Methods Exosomes were isolated from human DPSCs cultured undergrowth and odontogenic differentiation conditions, named UN-Exo and OD-Exo, respectively. The microRNA sequencing was performed to explore the microRNA profile contained in UN-Exo and OD-Exo. Pathway analysis was taken to detect enriched pathways associated with the predicted target genes of microRNAs. The regulatory roles of a highly expressed microRNA in OD-Exo were investigated through its inhibition or overexpression (miRNA inhibitors and miRNA mimics). Automated western blot was used to identify the function of exosomal microRNA and the roles of TGFβ1/smads pathway in odontogenic differentiation of DPSCs. A luciferase reporter gene assay was used to verify the direct target gene of exosomal miR-27a-5p. Results Endocytosis of OD-Exo triggered odontogenic differentiation of DPSCs by upregulating DSP, DMP-1, ALP, and RUNX2 proteins. MicroRNA sequencing showed that 28 microRNAs significantly changed in OD-Exo, of which 7 increased and 21 decreased. Pathway analysis showed genes targeted by differentially expressed microRNAs were involved in multiple signal transductions, including TGFβ pathway. 16 genes targeted by 15 differentially expressed microRNAs were involved in TGFβ signaling. Consistently, automated western blot found that OD-Exo activated TGFβ1 pathway by upregulating TGFβ1, TGFR1, p-Smad2/3, and Smad4 in DPSCs. Accordingly, once the TGFβ1 signaling pathway was inhibited by SB525334, protein levels of p-Smad2/3, DSP, and DMP-1 were significantly decreased in DPSCs treated with OD-Exo. MiR-27a-5p was expressed 11 times higher in OD-Exo, while miR-27a-5p promoted odontogenic differentiation of DPSCs and significantly upregulated TGFβ1, TGFR1, p-Smad2/3, and Smad4 by downregulating the inhibitory molecule LTBP1. Conclusions The microRNA expression profiles of exosomes derived from DPSCs were identified. OD-Exo isolated under odontogenic conditions were better inducers of DPSC differentiation. Exosomal microRNAs promoted odontogenic differentiation via TGFβ1/smads signaling pathway by downregulating LTBP1. Electronic supplementary material The online version of this article (10.1186/s13287-019-1278-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoli Hu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China. .,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Yingqun Zhong
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Kong
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Endodontics, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanan Chen
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junming Feng
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianmao Zheng
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China. .,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|