101
|
Li D, Niu G, Landén NX. Beyond the Code: Noncoding RNAs in Skin Wound Healing. Cold Spring Harb Perspect Biol 2022; 14:a041230. [PMID: 35197246 PMCID: PMC9438779 DOI: 10.1101/cshperspect.a041230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
An increasing number of noncoding RNAs (ncRNAs) have been found to regulate gene expression and protein functions, playing important roles in diverse biological processes and diseases. Their crucial functions have been reported in almost every cell type and all stages of skin wound healing. Evidence of their pathogenetic roles in common wound complications, such as chronic nonhealing wounds and excessive scarring, is also accumulating. Given their unique expression and functional properties, ncRNAs are promising therapeutic and diagnostic entities. In this review, we discuss current knowledge about the functional roles of noncoding elements, such as microRNAs, long ncRNAs, and circular RNAs, in skin wound healing, focusing on in vivo evidence from studies of human wound samples and animal wound models. Finally, we provide a perspective on the outlook of ncRNA-based therapeutics in wound care.
Collapse
Affiliation(s)
- Dongqing Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Guanglin Niu
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Ning Xu Landén
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institute, 17177 Stockholm, Sweden
| |
Collapse
|
102
|
Ren H, Zhao F, Zhang Q, Huang X, Wang Z. Autophagy and skin wound healing. BURNS & TRAUMA 2022; 10:tkac003. [PMID: 35187180 PMCID: PMC8847901 DOI: 10.1093/burnst/tkac003] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/07/2022] [Indexed: 02/07/2023]
Abstract
Autophagy is a lysosome-dependent, self-renewal mechanism that can degrade and recycle cellular components in eukaryotic cells to maintain the stability of the intracellular environment and the cells ability to cope with unfavorable environments. Numerous studies suggest that autophagy participates in regulating various cellular functions and is closely associated with the onset and progression of various diseases. Wound healing is a complex, multistep biological process that involves multiple cell types. Refractory wounds, which include diabetic skin ulcers, can seriously endanger human health. Previous studies have confirmed that autophagy plays an essential role in various phases of wound healing. Specifically, in the inflammatory phase, autophagy has an anti-infection effect and it negatively regulates the inflammatory response, which prevents excessive inflammation from causing tissue damage. In the proliferative phase, local hypoxia in the wound can induce autophagy, which plays a role in anti-apoptosis and anti-oxidative stress and promotes cell survival. Autophagy of vascular endothelial cells promotes wound angiogenesis and that of keratinocytes promotes their differentiation, proliferation and migration, which is conducive to the completion of wound re-epithelialisation. In the remodeling phase, autophagy of fibroblasts affects the formation of hypertrophic scars. Additionally, a refractory diabetic wound may be associated with increased levels of autophagy, and the regulation of mesenchymal stem cell autophagy may improve its application to wound healing. Therefore, understanding the relationship between autophagy and skin wound healing and exploring the molecular mechanism of autophagy regulation may provide novel strategies for the clinical treatment of wound healing.
Collapse
Affiliation(s)
- Haiyue Ren
- Department of Pathology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China
| | - Qiqi Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| | - Xing Huang
- Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| |
Collapse
|
103
|
Li M, Ding W, Liu G, Wang J. Extracellular Circular RNAs Act as Novel First Messengers Mediating Cell Cross-Talk in Ischemic Cardiac Injury and Myocardial Remodeling. J Cardiovasc Transl Res 2022; 15:444-455. [PMID: 35182317 DOI: 10.1007/s12265-022-10219-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
Abstract
Myocardial infarction (MI) causes most of the mortality worldwide. Coronary obstruction-caused myocardial ischemic injury leads to permanent loss of the myocardium. Subsequent compensatory myocardial remodeling and heart failure would result in arrhythmia and even sudden death. The molecular mechanisms of these pathological processes remain to be thoroughly revealed. Circular RNAs (circRNAs) are special types of non-coding RNAs which can durably regulate gene expression and modulate cell fate. They had been reported to mediate ischemic myocardial injury and myocardial remodeling. circRNAs can be loaded into extracellular vesicles and released into extracellular space. More recently, it was uncovered that the extracellular circRNAs can regulate intercellular communications, similar to "first messengers." The cross-talk mediated by extracellular circRNAs had been demonstrated to play important roles in pathological processes. Here, we would like to review the modulation of extracellular circRNAs in ischemic myocardial injury and myocardial remodeling. We believe the extracellular circRNAs can bring new strategies of MI treatment.
Collapse
Affiliation(s)
- Mengyang Li
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China
| | - Wei Ding
- Department of Comprehensive Internal Medicine, Affiliated Hospital, Qingdao University, Qingdao, 266003, China.
| | - Gaoli Liu
- Department of Cardiovascular Surgery, Affiliated Hospital, Qingdao University, Qingdao, 266510, China
| | - Jianxun Wang
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
104
|
Mahmoudi A, Butler AE, Jamialahmadi T, Sahebkar A. The role of exosomal miRNA in nonalcoholic fatty liver disease. J Cell Physiol 2022; 237:2078-2094. [PMID: 35137416 DOI: 10.1002/jcp.30699] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) impacts more than one-third of the population and is linked with other metabolic diseases. The term encompasses a wide spectrum of diseases, from modest steatosis to nonalcoholic steatohepatitis, fibrosis and, ultimately, cirrhosis with the potential for development of hepatocellular carcinoma. Currently, available methods for diagnosing NAFLD are invasive or lack accuracy, and monitoring to determine response to therapeutic interventions is challenging. Exosomes are nano-scaled extracellular vesicles that are secreted by a variety of cells. They convey proteins, mRNA, miRNA, and other bioactive molecules between cells and are involved in an extensive range of biological processes, particularly cell-cell communication. Several reports suggest that exosomes mediate miRNAs and, thus, they have potential clinical utility for diagnosis, prognosis, and therapeutics in liver diseases. In view of the vital role of exosomal microRNA in disease, we here synthesized current knowledge about the biogenesis of exosomal miRNA and exosome-mediated microRNA transfer. We then discuss the potential of exosomal miRNA in diagnosis and therapeutics of NAFLD.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Tannaz Jamialahmadi
- Surgical Oncology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
105
|
Xu YX, Pu SD, Li X, Yu ZW, Zhang YT, Tong XW, Shan YY, Gao XY. Exosomal ncRNAs: Novel Therapeutic Target and Biomarker for Diabetic Complications. Pharmacol Res 2022; 178:106135. [DOI: 10.1016/j.phrs.2022.106135] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 02/08/2023]
|
106
|
Chang W, Li M, Song L, Miao S, Yu W, Wang J. Noncoding RNAs from tissue-derived small extracellular vesicles: Roles in diabetes and diabetic complications. Mol Metab 2022; 58:101453. [PMID: 35121168 PMCID: PMC8866070 DOI: 10.1016/j.molmet.2022.101453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/22/2022] [Accepted: 01/28/2022] [Indexed: 12/01/2022] Open
Abstract
Diabetes is a systemic disease, and its progression involves multiple organ dysfunction. However, the exact mechanisms underlying pathological progression remain unclear. Small extracellular vesicles (sEVs) mediate physiological and pathological signaling communication between organs and have been shown to have important regulatory roles in diabetes and its complications in recent years. In particular, the majority of studies in the diabetes-related research field have focused on the noncoding RNAs carried by sEVs. Researchers found that noncoding RNA sorting into sEVs is not random but selective. Both tissue origin differences and environmental variations affect the cargo of sEVs. In addition, the function of sEVs differs according to the tissue they derive from; for example, sEVs derived from adipose tissue regulate insulin sensitivity in the periphery, while sEVs derived from bone marrow promote β-cell regeneration. Therefore, understanding the roles of sEVs from different tissues is important for elucidating their molecular mechanisms and is necessary for the application of sEVs as therapeutic agents for diabetes treatment in the future. In this review, we summarized current studies on the mechanisms of noncoding RNA sorting into sEVs, as well as the research progress on the effects of sEVs from different tissue origins and noncoding RNAs in diabetes and diabetic complications. The knowledge of noncoding RNAs in sEVs will help us better understand the role of sEVs in the diabetes progression.
Collapse
Affiliation(s)
- Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital, Qingdao University, Qingdao, China.
| | - Mengyang Li
- School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, China
| | - Lin Song
- School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, China
| | - Suo Miao
- School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, China
| | - Wanpeng Yu
- College of Medicine, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
107
|
Lu HJ, Li J, Yang G, Yi CJ, Zhang D, Yu F, Ma Z. Circular RNAs in stem cells: from basic research to clinical implications. Biosci Rep 2022; 42:BSR20212510. [PMID: 34908111 PMCID: PMC8738868 DOI: 10.1042/bsr20212510] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Circular RNAs (circRNAs) are a special class of endogenous RNAs with a wide variety of pathophysiological functions via diverse mechanisms, including transcription, microRNA (miRNA) sponge, protein sponge/decoy, and translation. Stem cells are pluripotent cells with unique properties of self-renewal and differentiation. Dysregulated circRNAs identified in various stem cell types can affect stem cell self-renewal and differentiation potential by manipulating stemness. However, the emerging roles of circRNAs in stem cells remain largely unknown. This review summarizes the major functions and mechanisms of action of circRNAs in stem cell biology and disease progression. We also highlight circRNA-mediated common pathways in diverse stem cell types and discuss their diagnostic significance with respect to stem cell-based therapy.
Collapse
Affiliation(s)
- Hui-Juan Lu
- The First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, China
| | - Juan Li
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guodong Yang
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, Hubei 438000, China
| | - Cun-Jian Yi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, China
| | - Daping Zhang
- The First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
| | - Fenggang Yu
- Institute of Life Science, Yinfeng Biological Group, Jinan 250000, China
| | - Zhaowu Ma
- The First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
| |
Collapse
|
108
|
Wu X, Jin S, Ding C, Wang Y, He D, Liu Y. Mesenchymal Stem Cell-Derived Exosome Therapy of Microbial Diseases: From Bench to Bed. Front Microbiol 2022; 12:804813. [PMID: 35046923 PMCID: PMC8761948 DOI: 10.3389/fmicb.2021.804813] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Microbial diseases are a global health threat, leading to tremendous casualties and economic losses. The strategy to treat microbial diseases falls into two broad categories: pathogen-directed therapy (PDT) and host-directed therapy (HDT). As the typical PDT, antibiotics or antiviral drugs directly attack bacteria or viruses through discerning specific molecules. However, drug abuse could result in antimicrobial resistance and increase infectious disease morbidity. Recently, the exosome therapy, as a HDT, has attracted extensive attentions for its potential in limiting infectious complications and targeted drug delivery. Mesenchymal stem cell-derived exosomes (MSC-Exos) are the most broadly investigated. In this review, we mainly focus on the development and recent advances of the application of MSC-Exos on microbial diseases. The review starts with the difficulties and current strategies in antimicrobial treatments, followed by a comprehensive overview of exosomes in aspect of isolation, identification, contents, and applications. Then, the underlying mechanisms of the MSC-Exo therapy in microbial diseases are discussed in depth, mainly including immunomodulation, repression of excessive inflammation, and promotion of tissue regeneration. In addition, we highlight the latest progress in the clinical translation of the MSC-Exo therapy, by summarizing related clinical trials, routes of administration, and exosome modifications. This review will provide fundamental insights and future perspectives on MSC-Exo therapy in microbial diseases from bench to bedside.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology and National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
109
|
Yang M, Chen J, Chen L. The roles of mesenchymal stem cell-derived exosomes in diabetes mellitus and its related complications. Front Endocrinol (Lausanne) 2022; 13:1027686. [PMID: 36339446 PMCID: PMC9633677 DOI: 10.3389/fendo.2022.1027686] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetes mellitus is a type of metabolic disease characterized by hyperglycemia, primarily caused by defects in insulin secretion, insulin action, or both. Long-term chronic hyperglycemia can lead to diabetes-related complications, causing damage, dysfunction, and failure of different organs. However, traditional insulin and oral drug therapy can only treat the symptoms but not delay the progressive failure of pancreatic beta cells or prevent the emergence of diabetic complications. Mesenchymal stem cells have received extensive attention due to their strong immunoregulatory functions and regeneration effects. Mesenchymal stem cell-derived exosomes (MSC-Exos) have been proposed as a novel treatment for diabetic patients as they have demonstrated superior efficiency to mesenchymal stem cells. This review summarizes the therapeutic effects, mechanisms, challenges, and future prospects of MSC-Exos in treating diabetes mellitus and its related complications. This review supports the potential use of MSC-Exos in future regenerative medicine to overcome the current difficulties in clinical treatment, particularly in treating diabetes.
Collapse
Affiliation(s)
- Mengmeng Yang
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
| | - Jun Chen
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, China
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, China
- Jinan Clinical Research Center for Endocrine and Metabolic Diseases, Jinan, China
- *Correspondence: Jun Chen, ; Li Chen,
| | - Li Chen
- Department of Endocrinology, Qilu Hospital, Shandong University, Jinan, China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, China
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, China
- Jinan Clinical Research Center for Endocrine and Metabolic Diseases, Jinan, China
- *Correspondence: Jun Chen, ; Li Chen,
| |
Collapse
|
110
|
Cao M, Bu C, Zhang J, Ren Y, Zhou G, Chen C, Han G, Jiang SW, Wen J. Exosomal Circular RNA hsa_circ_0046060 of Umbilical Cord Mesenchymal Stromal Cell Ameliorates Glucose Metabolism and Insulin Resistance in Gestational Diabetes Mellitus via the miR-338-3p/G6PC2 Axis. Int J Endocrinol 2022; 2022:9218113. [PMID: 35726320 PMCID: PMC9206588 DOI: 10.1155/2022/9218113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Impaired glucose metabolism and insulin sensitivity have been linked to the pathogenesis of gestational diabetes mellitus (GDM). Exosomes secreted by the umbilical cord mesenchymal stromal cells (UMSCs) and circular RNAs (circRNAs) derived from exosomes have been shown to be associated with the progression of GDM-related complications. METHODS UMSCs were isolated from umbilical cords and identified through flow cytometry. Exosomes were isolated from UMSCs and were then characterized. The expression levels of RNA of hsa_circ_0046060, mmu_circ_0002819, and miR-338-3p were determined by quantitative real-time polymerase chain reaction (RT-qPCR). The intracellular glucose intake and glycogen content were measured using a High Sensitivity Glucose Assay Kit and Glycogen Assay Kit, respectively. Bioinformatics analysis and luciferase reporter assay were used to validate interactions among hsa_circ_0046060, miR-338-3p, and G6PC2. The expression of insulin receptor substrate-1 (IRS-1) and its phosphorylated form, (p-IRS-1), as well as G6PC2, was determined through western blotting. RESULTS UMSCs and exosomes were successfully isolated and identified. The upregulation of hsa_circ_0046060 decreased the intracellular glucose content in L-02 cells (43.45 vs. 16.87 pM/mg, P=0.0002), whereas shRNA-mediated downregulation reversed this effect (16.87 vs. 33.16 pM/mg, P=0.0011). Mmu_circ_0002819 in mice aggravated dysregulated glucose metabolism (49.88 vs. 21.69 pM/mg, P=0.0031) and insulin sensitivity (0.20 vs. 0.11 mg/mL, P=0.03) in GDM mice, which was abrogated by the knockdown of mmu_circ_0002819. The results of luciferase reporter assay revealed that miR-338-3p and G6PC2 were the potential targets of has_circ_0046060. Western blotting results showed that the reduced activation of IRS-1 induced by GDM (1.25 vs. 0.54, P=0.0001) could be rescued by the administration of si-circ-G-UMSC-EXOs (0.54 vs. 1.17, P=0.0001). CONCLUSION Taken together, the inhibition of hsa_circ_0046060 expression in exosomes from GDM-derived UMSCs can alleviate GDM by reversing abnormal glucose metabolism and insulin resistance in vivo and in vitro.
Collapse
Affiliation(s)
- Minkai Cao
- Department of Gynecology and Obstetrics, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
- Department of Obstetrics and Gynecology, The Affiliated Wuxi Matemity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, Jiangsu, China
| | - Chaozhi Bu
- Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Matemity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, Jiangsu, China
| | - Jingjing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Yongwei Ren
- Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Matemity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, Jiangsu, China
| | - Guanlun Zhou
- Department of Gynecology and Obstetrics, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Chao Chen
- Department of Gynecology and Obstetrics, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Guorong Han
- Department of Gynecology and Obstetrics, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Shi-Wen Jiang
- Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Matemity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, Jiangsu, China
| | - Juan Wen
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| |
Collapse
|
111
|
Deng H, Chen Y. The role of adipose-derived stem cells-derived extracellular vesicles in the treatment of diabetic foot ulcer: Trends and prospects. Front Endocrinol (Lausanne) 2022; 13:902130. [PMID: 35966086 PMCID: PMC9363610 DOI: 10.3389/fendo.2022.902130] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic foot ulcer(DFU) is one of the most severe chronic complications of type 2 diabetes mellitus, which is mainly caused by peripheral vascular occlusion with various degrees of infection. Treatment of DFU is difficult, and ulcer formation in lower limbs and deep-tissue necrosis might lead to disability or even death. Insulin resistance is the major mechanism of type 2 diabetes mellitus development, largely caused by adipose tissue dysfunction. However, adipose tissue was recently identified as an important endocrine organ that secretes bio-active factors, such as adipokines and extracellular vesicles(EVs). And adipose tissue-derived stem cells(ADSCs) are abundant in adipose tissue and have become a hot topic in the tissue engineering field. In particular, EVs derived from ADSCs contain abundant biomarkers and mediators. These EVs exert significant effects on distant cells and organs, contributing to metabolic homeostasis. In this review, we aim to elaborate on the mechanisms of diabetic non-healing wound development and the role of ADSCs-EVs in wound repair, which might provide a new therapy for treating DFU.
Collapse
Affiliation(s)
- Hongyan Deng
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
- *Correspondence: Yong Chen,
| |
Collapse
|
112
|
Han ZF, Cao JH, Liu ZY, Yang Z, Qi RX, Xu HL. Exosomal lncRNA KLF3-AS1 derived from bone marrow mesenchymal stem cells stimulates angiogenesis to promote diabetic cutaneous wound healing. Diabetes Res Clin Pract 2022; 183:109126. [PMID: 34742784 DOI: 10.1016/j.diabres.2021.109126] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023]
Abstract
AIMS We focused on BMSC-derived exosomal lncRNA KLF3-AS1 and its significance in diabetic cutaneous wound healing. METHODS Potential interaction between KLF3-AS1 and miR-383, miR-383 and VEGFA were predicted using bioinformatic analysis and validated by luciferase reporter, RIP, and FISH assays. The proliferation, apoptosis, migration and tube formation of HUVECs were evaluated by CCK-8, flow cytometry, wound healing, and tube formation assays, respectively. A murine diabetic cutaneous wound model was used to investigate therapeutic effects of exosomal KLF3-AS1 in vivo. Histological alterations in skin tissues were examined using HE, Masson staining, and immunostaining of CD31. RESULTS BMSC-derived exosomal KLF3-AS1 sufficiently promoted proliferation, migration, and tube formation, while inhibited apoptosis of HUVECs challenged by high glucose. The protective effects of exosomal KLF3-AS1 were achieved at least partially by down-regulating miR-383, and boosting the expression of its target, VEGFA. In vivo, exosomes from KLF3-AS1-expressing BMSCs demonstrated the best effects in promoting cutaneous wound healing in diabetic mice, which were associated with minimal weight loss, increased blood vessel formation, reduced inflammation, decreased miR-383 expression, and up-regulated VEGFA. CONCLUSIONS Exosomal lncRNA KLF3-AS1 derived from BMSCs induces angiogenesis to promote diabetic cutaneous wound healing.
Collapse
Affiliation(s)
- Zhao-Feng Han
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, China.
| | - Jun-Hua Cao
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, China
| | - Zhong-Yang Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, China
| | - Zheng Yang
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, China
| | - Rui-Xue Qi
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, China
| | - Hua-Lin Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, China
| |
Collapse
|
113
|
Molibeli KM, Hu R, Liu Y, Xiong D, Tang L. Potential Clinical Applications of Exosomal Circular RNAs: More than Diagnosis. Front Mol Biosci 2021; 8:769832. [PMID: 34901159 PMCID: PMC8652074 DOI: 10.3389/fmolb.2021.769832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/04/2021] [Indexed: 12/20/2022] Open
Abstract
Exosomes are small vesicles derived from cells used as cell-to-cell communication goods in numerous diseases including tumorigenesis, neurological diseases, cardiovascular diseases and other diseases. Circular RNAs (circRNAs) are an innovative constituent of non-coding endogenous RNAs generated through backsplicing, catalyzed by RNA polymerase Ⅱ. These non-coding RNAs have been suggested to control gene expression through miRNA sponging, RNA-binding protein regulation and translational capabilities. Genome-wide RNA sequence analyses observed that circRNAs were stably improved in exosomes in association to parental cells. Little attention has been dedicated to exosomal circRNAs (exo-circRNAs). However, research has demonstrated that exo-circRNAs may have important regulatory functions because of their stability in cells and within exosomes. If well understood, the precise roles and mechanisms of exo-circRNAs might surge the impending clinical applications of these molecules as markers in the identification, prediction and treatment of various diseases. In this review, we outline recent findings regarding exo-circRNAs which includes their functions and highlights their potential applications and therapeutic targets in human diseases.
Collapse
Affiliation(s)
- Kearabetsoe Matseliso Molibeli
- School of Life Science, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Rong Hu
- School of Life Science, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuze Liu
- School of Life Science, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Dehui Xiong
- School of Life Science, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Lijun Tang
- School of Life Science, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
114
|
Jiang LN, Ji X, Liu W, Qi C, Zhai X. Identification of the circ_PRKDC/miR-20a-3p/RASA1 axis in regulating HaCaT keratinocyte migration. Wound Repair Regen 2021; 30:282-291. [PMID: 34897876 DOI: 10.1111/wrr.12988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/20/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022]
Abstract
Migration of keratinocytes plays a crucial role in the re-epithelialization phase during wound healing. Circular RNA (circRNA) protein kinase, DNA-activated, catalytic subunit (circ_PRKDC, hsa_circ_0084443) has been identified as a regulator of keratinocyte migration. However, the molecular basis governing it remains unclear. The levels of circ_PRKDC, microRNA (miR)-20a-3p, and RAS p21 protein activator 1 (RASA1) were assessed by quantitative real-time PCR (qRT-PCR) or western blot. Subcellular localization, Actinomycin D, and Ribonuclease (RNase) R assays were performed to characterise circ_PRKDC. Cell migration was gauged by transwell and wound-healing assays. A direct relationship between miR-20a-3p and circ_PRKDC or RASA1 was verified by dual-luciferase reporter and RNA pull-down assays. Circ_PRKDC expression was reduced in wound skin during wound healing. Circ_PRKDC modulated migration of HaCaT keratinocytes. Mechanistically, circ_PRKDC directly targeted miR-20a-3p. The regulation of circ_PRKDC on HaCaT keratinocyte migration was mediated by miR-20a-3p. RASA1 was identified as a direct and functional target of miR-20a-3p, and miR-20a-3p-mediated inhibition of RASA1 impacted HaCaT keratinocyte migration. Circ_PRKDC acted as a post-transcriptional modulator of RASA1 expression through miR-20a-3p. Moreover, circ_PRKDC modulated migration of HaCaT keratinocytes by RASA1. Our findings demonstrated a novel molecular basis, the miR-20a-3p/RASA1 axis, for the regulation of circ_PRKDC on HaCaT keratinocyte migration.
Collapse
Affiliation(s)
- Li-Na Jiang
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaohui Ji
- Department of Pathology, The People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Wei Liu
- Department of Breast Surgery, The People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Chuanchuan Qi
- Department of Breast Surgery, The People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Xiaomei Zhai
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
115
|
Hong P, Yu M, Tian W. Diverse RNAs in adipose-derived extracellular vesicles and their therapeutic potential. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:665-677. [PMID: 34703651 PMCID: PMC8516999 DOI: 10.1016/j.omtn.2021.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adipose tissue, which is considered an energy storage and active endocrine organ, produces and secretes a large amount of adipokines to regulate distant targets through blood circulation, especially extracellular vesicles (EVs). As cell-derived, membranous nanoparticles, EVs have recently garnered great attention as novel mediators in establishing intercellular communications as well as in accelerating interorgan crosstalk. Studies have revealed that the RNAs, including coding RNAs (messenger RNAs) and noncoding RNAs (long noncoding RNAs, microRNAs, and circular RNAs) are key bioactive cargoes of EV functions in various pathophysiological processes, such as cell differentiation, metabolic homeostasis, immune signal transduction, and cancer. Moreover, certain EV-contained RNAs have gradually been recognized as novel biomarkers, prognostic indicators, or even therapeutic nanodrugs of diseases. Therefore, in this review, we comprehensively summarize different classes of RNAs presented in adipose-derived EVs and discuss their therapeutic potential according to the latest research progress to provide valuable knowledge in this area.
Collapse
Affiliation(s)
- Pengyu Hong
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mei Yu
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases West China School of Stomatology, Sichuan University, Chengdu, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
116
|
Potential Applications and Functional Roles of Exosomes in Cardiometabolic Disease. Pharmaceutics 2021; 13:pharmaceutics13122056. [PMID: 34959338 PMCID: PMC8703910 DOI: 10.3390/pharmaceutics13122056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Despite diagnostic and therapeutic advances, cardiometabolic disease remains the leading cause of death worldwide. Extracellular vesicles (EVs), which include exosomes and microvesicles, have gained particular interest because of their role in metabolic homeostasis and cardiovascular physiology. Indeed, EVs are recognized as critical mediators of intercellular communication in the cardiovascular system. Exosomes are naturally occurring nanocarriers that transfer biological information in the setting of metabolic abnormalities and cardiac dysfunction. The study of these EVs can increase our knowledge on the pathophysiological mechanisms of metabolic disorders and their cardiovascular complications. Because of their inherent properties and composition, exosomes have been proposed as diagnostic and prognostic biomarkers and therapeutics for specific targeting and drug delivery. Emerging fields of study explore the use exosomes as tools for gene therapy and as a cell-free alternative for regenerative medicine. Furthermore, innovative biomaterials can incorporate exosomes to enhance tissue regeneration and engineering. In this work, we summarize the most recent knowledge on the role of exosomes in cardiometabolic pathophysiology while highlighting their potential therapeutic applications.
Collapse
|
117
|
Abstract
Exosomes are nano-sized extracellular vesicles (30–160 nm diameter) with lipid bilayer membrane secrete by various cells that mediate the communication between cells and tissue, which contain a variety of non-coding RNAs, mRNAs, proteins, lipids and other functional substances. Adipose tissue is important energy storage and endocrine organ in the organism. Recent studies have revealed that adipose tissue-derived exosomes (AT-Exosomes) play a critical role in many physiologically and pathologically functions. Physiologically, AT-Exosomes could regulate the metabolic homoeostasis of various organs or cells including liver and skeletal muscle. Pathologically, they could be used in the treatment of disease and or that they may be involved in the progression of the disease. In this review, we describe the basic principles and methods of exosomes isolation and identification, as well as further summary the specific methods. Moreover, we categorize the relevant studies of AT-Exosomes and summarize the different components and biological functions of mammalian exosomes. Most importantly, we elaborate AT-Exosomes crosstalk within adipose tissue and their functions on other tissues or organs from the physiological and pathological perspective. Based on the above analysis, we discuss what remains to be discovered problems in AT-Exosomes studies and prospect their directions needed to be further explored in the future.
Collapse
Affiliation(s)
- Rui Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Tiantian Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Zhaozhao He
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Rui Cai
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&f University, Yangling, China
| |
Collapse
|
118
|
Circ_0084443 Inhibits Wound Healing Via Repressing Keratinocyte Migration Through Targeting the miR-17-3p/FOXO4 Axis. Biochem Genet 2021; 60:1236-1252. [PMID: 34837127 DOI: 10.1007/s10528-021-10157-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Abstract
Keratinocyte migration is a crucial process during skin wound healing, and circular RNAs are associated with keratinocyte migration. The purpose of our study was to clarify the role of circ_0084443 in wound healing. The levels of circ_0084443, microRNA (miR)-17-3p, and forkhead box protein O4 (FOXO4) were examined by quantitative reverse transcription-PCR. Cell migration was detected via wound scratch assay or transwell assay. The protein expression was measured using western blot. The binding analysis between miR-17-3p and circ_0084443 or FOXO4 was determined by dual-luciferase reporter assay and RNA Immunoprecipitation assay. TGF-β1 decreased the levels of circ_0084443 and FOXO4 while increased the miR-17-3p expression in keratinocytes by a concentration-dependent manner. Circ_0084443 acted as a miR-17-3p sponge and circ_0084443 overexpression alleviated TGF-β1-induced migration of keratinocytes by sponging miR-17-3p. FOXO4 was a target for miR-17-3p. The downregulation of miR-17-3p suppressed cell migration in TGF-β1-induced cells by increasing the FOXO4 level. Circ_0084443 positively regulated the FOXO4 expression by sponging miR-17-3p. Circ_0084443 suppressed the TGFβ signaling pathway by affecting the miR-17-3p/FOXO4 axis. These results exhibited that circ_0084443 suppressed the TGF-β1-induced keratinocyte migration by regulating the miR-17-3p/FOXO4 axis, suggesting the application potential of circ_0084443 in wound-healing-related diseases.
Collapse
|
119
|
Roles and mechanisms of exosomal non-coding RNAs in human health and diseases. Signal Transduct Target Ther 2021; 6:383. [PMID: 34753929 PMCID: PMC8578673 DOI: 10.1038/s41392-021-00779-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes play a role as mediators of cell-to-cell communication, thus exhibiting pleiotropic activities to homeostasis regulation. Exosomal non-coding RNAs (ncRNAs), mainly microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are closely related to a variety of biological and functional aspects of human health. When the exosomal ncRNAs undergo tissue-specific changes due to diverse internal or external disorders, they can cause tissue dysfunction, aging, and diseases. In this review, we comprehensively discuss the underlying regulatory mechanisms of exosomes in human diseases. In addition, we explore the current knowledge on the roles of exosomal miRNAs, lncRNAs, and circRNAs in human health and diseases, including cancers, metabolic diseases, neurodegenerative diseases, cardiovascular diseases, autoimmune diseases, and infectious diseases, to determine their potential implication in biomarker identification and therapeutic exploration.
Collapse
|
120
|
Shang B, Xu T, Hu N, Mao Y, Du X. Circ-Klhl8 overexpression increased the therapeutic effect of EPCs in diabetic wound healing via the miR-212-3p/SIRT5 axis. J Diabetes Complications 2021; 35:108020. [PMID: 34507876 DOI: 10.1016/j.jdiacomp.2021.108020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/15/2022]
Abstract
Previous studies found that hypoxic pretreatment of endothelial progenitor cells (EPCs) prior to transplantation had a greater therapeutic effect than untreated EPCs in promoting diabetic wound healing. However, the exact mechanism is uncertain. Here, circRNA expression in EPCs after hypoxic treatment was investigated. High-throughput sequencing was used to assess abnormal expression by EPCs of circular RNAs (circRNAs) following hypoxic pretreatment. Additionally, an in vivo full-thickness skin defect mouse model was used to assess the effects of transplanted EPCs on diabetic wound closure. Subsequently, the regulatory mechanism and targets were studied. The results showed that circ-Klhl8 overexpression suppressed hyper glucose-induced endothelial cell damage by activating autophagy. MiR-212-3p and SIRT5 were identified as the downstream targets of circ-Klhl8. Circ-Klhl8 overexpression promoted skin wound healing by regulating SIRT5-mediated autophagy. In conclusion, the study found that circ-Klhl8 overexpression increased the EPC therapeutic effect in promoting diabetic wound healing by targeting the miR-212-3p/SIRT5 axis.
Collapse
Affiliation(s)
- Bin Shang
- Department of Cardio-Thoracic Surgery, The Second Affiliated Hospital of BBMC, #633 Longhua Road, Huaishang District, Bengbu, 233000 Anhui, China
| | - Tianze Xu
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008 Jiangsu, China
| | - Nan Hu
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008 Jiangsu, China.
| | - Youjun Mao
- Department of Vascular Surgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, #68 Gehu Zhong Road, Wujin District, Changzhou City 213000, China.
| | - Xiaolong Du
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008 Jiangsu, China.
| |
Collapse
|
121
|
Wang SZ, Jia J, Chen CH. lncRNA-KCNQ1OT1: A Potential Target in Exosomes Derived from Adipose-Derived Stem Cells for the Treatment of Osteoporosis. Stem Cells Int 2021; 2021:7690006. [PMID: 34712334 PMCID: PMC8548139 DOI: 10.1155/2021/7690006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Osteoporosis is a worldwide medical and socioeconomic burden characterized by systemic impairment of bone strength and microstructure. Exosomes derived from adipose-derived stem cells (ADSCs-Exos) have been confirmed to play effective roles in the repair of various tissues and organs. This study was aimed at investigating the role of ADSCs-Exos and a novel long noncoding RNA KCNQ1OT1 played in osteoporosis as well as the underlying mechanism. METHODS Primary osteoblasts were treated with different doses of tumor necrosis factor-α (TNF-α) (0, 1, 2.5, 5, and 10 ng/ml) and then cocultured with ADSCs-Exos or exosome-derived from lnc-KCNQ1OT1-modified ADSCs (KCNQ1OT1-Exos). The expression of miRNA-141-5p (miR-141-5p) and lnc-KCNQ1OT1 was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). The protein expression of cleaved-caspase-3, caspase-3, and Bax was determined by Western blot. Cell viability and apoptosis were assessed by Cell Counting Kit-8 (CCK-8) and flow cytometry analysis, respectively. The binding sites between KCNQ1OT1 and miR-141-5p were validated by dual-luciferase reporter assay. RESULTS TNF-α dose-dependently increased miR-141-5p expression, inhibited viability, and promoted apoptosis of osteoblasts. However, miR-141-5p silencing or cocultured with ADSCs-Exos attenuated these effects. In addition, KCNQ1OT1-Exos could more significantly attenuate the induced cytotoxicity and apoptosis compared to ADSCs-Exos. Moreover, miR-141-5p was confirmed as the target of KCNQ1OT1 by luciferase reporter assay. CONCLUSIONS ADSCs-Exos can attenuate cytotoxicity and apoptosis of TNF-α-induced primary osteoblasts. KCNQ1OT1-Exos have a more significant inhibitory effect compared to ADSCs-Exos by the function of sponging miR-141-5p, suggesting that KCNQ1OT1-Exos can be promising agents in osteoporosis treatment.
Collapse
Affiliation(s)
- Shan-Zheng Wang
- Department of Orthopaedics, Zhongda Hospital, Medical School of Southeast University, 87 Ding Jia Qiao Road, Nanjing, Jiangsu 210009, China
| | - Jun Jia
- Department of Orthopaedics, The 904th Hospital of Joint Logistic Support Force, PLA, 101 Xingyuan North Road, Wuxi, Jiangsu 214000, China
| | - Chang-Hong Chen
- Department of Orthopaedics, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, 130 Renmin Middle Road, Jiangyin, Jiangsu 214400, China
| |
Collapse
|
122
|
Li X, Li N, Li B, Feng Y, Zhou D, Chen G. Noncoding RNAs and RNA-binding proteins in diabetic wound healing. Bioorg Med Chem Lett 2021; 50:128311. [PMID: 34438011 DOI: 10.1016/j.bmcl.2021.128311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Poor wound healing is a common complication in diabetic patients. It often leads to intractable infections and lower limb amputations and is associated with cardiovascular morbidity and mortality. NcRNAs, which can regulate gene expression, have emerged as important regulators of various physiological processes. Herein, we summarize the diverse roles of ncRNAs in the key stages of diabetic wound healing, including inflammation, angiogenesis, re-epithelialization, and extracellular matrix remodeling. Meanwhile, the potential use of ncRNAs as novel therapeutic targets for wound healing in diabetic patients is also discussed. In addition, we summarize the role of RNA-binding proteins (RBPs) in the regulation of gene expression and signaling pathways during skin repair, which may provide opportunities for therapeutic intervention for this potentially devastating disease. However, so far, research on the modulated drug based on ncRNAs that lead to significantly altered gene expression in diabetic patients is scarce. We have compiled some drugs that may be able to modulate ncRNAs, which significantly regulate the gene expression in diabetic patients.
Collapse
Affiliation(s)
- Xue Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Bingxin Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yuan Feng
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Di Zhou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Gang Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China; Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, People's Republic of China.
| |
Collapse
|
123
|
Angiogenic Effects and Crosstalk of Adipose-Derived Mesenchymal Stem/Stromal Cells and Their Extracellular Vesicles with Endothelial Cells. Int J Mol Sci 2021; 22:ijms221910890. [PMID: 34639228 PMCID: PMC8509224 DOI: 10.3390/ijms221910890] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived mesenchymal stem/stromal cells (ASCs) are an adult stem cell population able to self-renew and differentiate into numerous cell lineages. ASCs provide a promising future for therapeutic angiogenesis due to their ability to promote blood vessel formation. Specifically, their ability to differentiate into endothelial cells (ECs) and pericyte-like cells and to secrete angiogenesis-promoting growth factors and extracellular vesicles (EVs) makes them an ideal option in cell therapy and in regenerative medicine in conditions including tissue ischemia. In recent angiogenesis research, ASCs have often been co-cultured with an endothelial cell (EC) type in order to form mature vessel-like networks in specific culture conditions. In this review, we introduce co-culture systems and co-transplantation studies between ASCs and ECs. In co-cultures, the cells communicate via direct cell-cell contact or via paracrine signaling. Most often, ASCs are found in the perivascular niche lining the vessels, where they stabilize the vascular structures and express common pericyte surface proteins. In co-cultures, ASCs modulate endothelial cells and induce angiogenesis by promoting tube formation, partly via secretion of EVs. In vivo co-transplantation of ASCs and ECs showed improved formation of functional vessels over a single cell type transplantation. Adipose tissue as a cell source for both mesenchymal stem cells and ECs for co-transplantation serves as a prominent option for therapeutic angiogenesis and blood perfusion in vivo.
Collapse
|
124
|
Zhang X, Han C. Bone Marrow Mesenchymal Stem Cell (BMSC)-Derived Exosomes Increase Colon Cancer Cell Apoptosis and Inhibit Proliferation and Migration. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study aims to investigate whether bone marrow mesenchymal stem cell (BMSC) exosomes (BMSC-exos) affects the progression of colon cancer. Ultracentrifugation was used to extract and collect BMSC-exos which were assessed under electron microscope and by flow cytometry. The BMSCs
were divided into two groups: control group treated with α-MEM basal medium and experimental group with exosomes (10 μg/ml). Exos were extracted from BMSCs and co-cultured with colon cancer cells, followed by analysis of cell viability by CCK-8 assay and GLUT3 mRNA and
protein expression by RT-qPCR and Western blot. The electron microscope analysis indicated that the primary BMSCs showed a long spindle shape with a negative expression of antigen CD34 and positive antigen CD90. Importantly, exos inhibited the viability of colon cancer cells HCT116 and decreased
the expression of GLUT3, suggesting that exos might increase the colon cancer cell apoptosis. In conclusion, BMSC-exos inhibit cell progression in colon cancer and might be served as a promising biomarker.
Collapse
Affiliation(s)
- Xinfa Zhang
- Department of General Surgery, Shandong Province Coal Taishan Sanatorium, Taian City, Shandong Province, 271000, China
| | - Cheng Han
- Department of General Surgery, Shandong Province Coal Taishan Sanatorium, Taian City, Shandong Province, 271000, China
| |
Collapse
|
125
|
Bray ER, Oropallo AR, Grande DA, Kirsner RS, Badiavas EV. Extracellular Vesicles as Therapeutic Tools for the Treatment of Chronic Wounds. Pharmaceutics 2021; 13:1543. [PMID: 34683836 PMCID: PMC8541217 DOI: 10.3390/pharmaceutics13101543] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic wounds develop when the orderly process of cutaneous wound healing is delayed or disrupted. Development of a chronic wound is associated with significant morbidity and financial burden to the individual and health-care system. Therefore, new therapeutic modalities are needed to address this serious condition. Mesenchymal stem cells (MSCs) promote skin repair, but their clinical use has been limited due to technical challenges. Extracellular vesicles (EVs) are particles released by cells that carry bioactive molecules (lipids, proteins, and nucleic acids) and regulate intercellular communication. EVs (exosomes, microvesicles, and apoptotic bodies) mediate key therapeutic effects of MSCs. In this review we examine the experimental data establishing a role for EVs in wound healing. Then, we explore techniques for designing EVs to function as a targeted drug delivery system and how EVs can be incorporated into biomaterials to produce a personalized wound dressing. Finally, we discuss the status of clinically deploying EVs as a therapeutic agent in wound care.
Collapse
Affiliation(s)
- Eric R. Bray
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alisha R. Oropallo
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Daniel A. Grande
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell Health, Hempstead, NY 11549, USA; (A.R.O.); (D.A.G.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
- Department of Orthopedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA
| | - Robert S. Kirsner
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
| | - Evangelos V. Badiavas
- Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.R.B.); (R.S.K.)
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
126
|
Girón J, Maurmann N, Pranke P. The role of stem cell-derived exosomes in the repair of cutaneous and bone tissue. J Cell Biochem 2021; 123:183-201. [PMID: 34514621 DOI: 10.1002/jcb.30144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022]
Abstract
Exosomes are extracellular vesicles secreted by various cell types, which play important roles in physiological processes. In particular, stem cell-derived exosomes have been shown to play crucial functions in intercellular communication during the tissue healing process. This review summarizes the effects of exosomes derived from different stem cell sources on the repair of cutaneous and bone tissue, focusing on the different pathways that could be involved in the regeneration process. The biogenesis, isolation, and content of exosomes have also been discussed. The effectiveness of exosomes is broadly demonstrated for skin and bone regeneration in animal models, supporting the basis for clinical translation of exosomes as a ready-to-use cell-free therapeutic for skin and bone regeneration.
Collapse
Affiliation(s)
- Juliana Girón
- Hematology & Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Post Graduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Natasha Maurmann
- Hematology & Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Post Graduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patricia Pranke
- Hematology & Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Post Graduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Stem Cell Research Institute, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
127
|
Guo J, Xiao F, Ren W, Zhu Y, Du Q, Li Q, Li X. Circular Ribonucleic Acid circFTO Promotes Angiogenesis and Impairs Blood-Retinal Barrier Via Targeting the miR-128-3p/Thioredoxin Interacting Protein Axis in Diabetic Retinopathy. Front Mol Biosci 2021; 8:685466. [PMID: 34422901 PMCID: PMC8371555 DOI: 10.3389/fmolb.2021.685466] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/08/2021] [Indexed: 01/16/2023] Open
Abstract
Background: Increasing attention has been attracted by the role of circular RNAs (circRNAs) in ocular diseases. Previous study has revealed that circ_0005941 (also known as circFTO, an alpha-ketoglutarate-dependent dioxygenase) was upregulated in the vitreous humor of diabetic retinopathy (DR), while its underlying mechanism in DR remains unknown. Methods: Retinal vascular endothelial cells (RVECs) treated with high glucose (HG) were used to establish the DR cell model. The in vivo assays were conducted using streptozotocin-induced diabetic mice. The circular structure and stability of circFTO were identified by Sanger sequencing and RNase R treatment. RT-qPCR analysis was used to detect the RNA expression. The levels of the mRNA-encoded protein thioredoxin-interacting protein (TXNIP) or angiogenesis-associated proteins (VEGFA, PDGF, and ANG2) and blood-retinal barrier (BRB)-related proteins (ZO-1, Occludin, and Claudin-5) were measured by Western blot. The viability of RVECs was measured using CCK-8 assays. The angiogenesis of RVECs was assessed using tube formation assays in vitro. Endothelial permeability assays were conducted to examine the function of the BRB. The binding between genes was explored using RNA pulldown and luciferase reporter assays. Results: CircFTO was upregulated in HG-treated RVECs. CircFTO deficiency reversed the HG-induced increase in the viability and angiogenesis of RVECs and alleviated HG-mediated impairment of the BRB. MiR-128-3p bound with circFTO and was downregulated in HG-treated RVECs. TXNIP was a downstream target gene of miR-128-3p. TXNIP was highly expressed in the DR cell model. Rescue assays revealed that circFTO promoted angiogenesis and impaired the blood-retinal barrier by upregulating TXNIP. In the DR mouse model, circFTO silencing inhibited angiogenesis and promoted BRB recovery in vivo. Conclusion: CircFTO promotes angiogenesis and impairs the blood-retinal barrier in vitro and in vivo by binding with miR-128-3p to upregulate TXNIP in DR.
Collapse
Affiliation(s)
- Jianjin Guo
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Feng Xiao
- Department of Oncology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Wei Ren
- Department of Endocrinology and Metabolism, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Yikun Zhu
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Qiujing Du
- Shanxi Medical University, Taiyuan, China
| | - Qian Li
- Shanxi Medical University, Jinzhong, China
| | - Xing Li
- Department of Endocrinology and Metabolism, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
128
|
Wu SC, Kuo PJ, Rau CS, Huang LH, Lin CW, Wu YC, Wu CJ, Tsai CW, Hsieh TM, Liu HT, Huang CY, Hsieh CH. Increased Angiogenesis by Exosomes Secreted by Adipose-Derived Stem Cells upon Lipopolysaccharide Stimulation. Int J Mol Sci 2021; 22:8877. [PMID: 34445582 PMCID: PMC8396299 DOI: 10.3390/ijms22168877] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/19/2022] Open
Abstract
Exosomes secreted by adipose-derived stem cells (ADSCs) enhance angiogenesis and wound healing. However, in clinical settings, wounds may be infected by various bacteria or pathogens. We investigated whether human ADSCs stimulated with lipopolysaccharide (LPS) secrete exosomes (ADSC-LPS-exo) that augment the angiogenesis of human umbilical vein endothelial cells (HUVECs). ExoQuick-TC exosome precipitation solution was used to purify exosomes from human ADSC culture media in the presence or absence of 1 µg/mL LPS treatment for 24 h. The uptake of ADSC-LPS-exo significantly induced the activation of cAMP response element binding protein (CREB), activating protein 1 (AP-1), and nuclear factor-κB (NF-κB) signaling pathways and increased the migration of and tube formation in HUVECs. RNA interference with CREB, AP-1, or NF-κB1 significantly reduced the migration of and tube formation in HUVECs treated with ADSC-LPS-exo. An experiment with an antibody array for 25 angiogenesis-related proteins revealed that only interleukin-8 expression was significantly upregulated in HUVECs treated with ADSC-LPS-exo. In addition, proteomic analysis revealed that eukaryotic translation initiation factor 4E, amyloid beta A4 protein, integrin beta-1, and ras-related C3 botulinum toxin substrate 1 may be potential candidates involved in ADSC-LPS-exo-mediated enhanced angiogenesis.
Collapse
Affiliation(s)
- Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan;
| | - Pao-Jen Kuo
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Lien-Hung Huang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Chia-Wei Lin
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Yi-Chan Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Jung Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Wen Tsai
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Ting-Min Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Hang-Tsung Liu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Chun-Ying Huang
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Ching-Hua Hsieh
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
129
|
Weiliang Z, Lili G. Research Advances in the Application of Adipose-Derived Stem Cells Derived Exosomes in Cutaneous Wound Healing. Ann Dermatol 2021; 33:309-317. [PMID: 34341631 PMCID: PMC8273313 DOI: 10.5021/ad.2021.33.4.309] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Cutaneous wound healing has always been an intractable medical problem for both clinicians and researchers, with an urgent need for more efficacious methods to achieve optimal outcomes morphologically and functionally. Stem cells, the body's rapid response 'road repair crew,' being on standby to combat tissue injuries, are an essential part of regenerative medicine. Currently, the use of adipose-derived stem cells (ADSCs), a kind of mesenchymal stem cells with multipotent differentiation and self-renewal capacity, is surging in the field of cutaneous wound healing. ADSCs may exert influences either by releasing paracrine signalling factors or differentiating into mature adipose cells to provide the 'building blocks' for engineered tissue. As an important paracrine substance released from ADSCs, exosomes are a kind of extracellular vesicles and carrying various bioactive molecules mediating adjacent or distant intercellular communication. Previous studies have indicated that ADSCs derived exosomes (ADSCs-Exos) promoted skin wound healing by affecting all stages of wound healing, including regulating inflammatory response, promoting proliferation and migration of fibroblasts or keratinocytes, facilitating angiogenesis, and regulating remodeling of extracellular matrix, which have provided new opportunities for understanding how ADSCs-Exos mediate intercellular communication in pathological processes of the skin and therapeutic strategies for cutaneous wound repair. In this review, we focus on elucidating the role of ADSCs-Exos at various stages of cutaneous wound healing, detailing the latest developments, and presenting some challenges necessary to be addressed in this field, with the expectation of providing a new perspective on how to best utilize this powerful cell-free therapy in the future.
Collapse
Affiliation(s)
- Zeng Weiliang
- Department of Cosmetic and Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo Lili
- Department of Cosmetic and Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
130
|
Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells 2021; 10:cells10081959. [PMID: 34440728 PMCID: PMC8393426 DOI: 10.3390/cells10081959] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a type of extracellular vesicles, produced within multivesicular bodies, that are then released into the extracellular space through a merging of the multivesicular body with the plasma membrane. These vesicles are secreted by almost all cell types to aid in a vast array of cellular functions, including intercellular communication, cell differentiation and proliferation, angiogenesis, stress response, and immune signaling. This ability to contribute to several distinct processes is due to the complexity of exosomes, as they carry a multitude of signaling moieties, including proteins, lipids, cell surface receptors, enzymes, cytokines, transcription factors, and nucleic acids. The favorable biological properties of exosomes including biocompatibility, stability, low toxicity, and proficient exchange of molecular cargos make exosomes prime candidates for tissue engineering and regenerative medicine. Exploring the functions and molecular payloads of exosomes can facilitate tissue regeneration therapies and provide mechanistic insight into paracrine modulation of cellular activities. In this review, we summarize the current knowledge of exosome biogenesis, composition, and isolation methods. We also discuss emerging healing properties of exosomes and exosomal cargos, such as microRNAs, in brain injuries, cardiovascular disease, and COVID-19 amongst others. Overall, this review highlights the burgeoning roles and potential applications of exosomes in regenerative medicine.
Collapse
|
131
|
He X, Kuang G, Wu Y, Ou C. Emerging roles of exosomal miRNAs in diabetes mellitus. Clin Transl Med 2021; 11:e468. [PMID: 34185424 PMCID: PMC8236118 DOI: 10.1002/ctm2.468] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Exosomes are small extracellular vesicles 40-160 nm in diameter that are secreted by almost all cell types. Exosomes can carry diverse cargo including RNA, DNA, lipids, proteins, and metabolites. Exosomes transfer substances and information between cells by circulating in body fluids and are thus involved in diverse physiological and pathological processes in the human body. Recent studies have closely associated exosomal microRNAs (miRNAs) with various human diseases, including diabetes mellitus (DM), which is a complex multifactorial metabolic disorder disease. Exosomal miRNAs are emerging as pivotal regulators in the progression of DM, mainly in terms of pancreatic β-cell injury and insulin resistance. Exosomal miRNAs are closely associated with DM-associated complications, such as diabetic retinopathy (DR), diabetic nephropathy (DN), and diabetic cardiomyopathy (DCM), etc. Further investigations of the mechanisms of action of exosomal miRNAs and their role in DM will be valuable for the thorough understanding of the physiopathological process of DM. Here, we have summarized recent findings regarding exosomal miRNAs associated with DM to provide a new strategy for identifying potential diagnostic biomarkers and drug targets for the early diagnosis and treatment, respectively, of DM.
Collapse
Affiliation(s)
- Xiaoyun He
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Departments of Ultrasound Imaging, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Gaoyan Kuang
- Department of OrthopedicsThe First Affiliated Hospital of Hunan University of Chinese MedicineChangshaHunan410007China
- Postdoctoral Research WorkstationHinye Pharmaceutical Co. LtdChangshaHunan410331China
| | - Yongrong Wu
- Hunan university of Chinese MedicineChangshaHunan410208China
| | - Chunlin Ou
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
132
|
Exosomes from Adipose Mesenchymal Stem Cells Overexpressing Stanniocalcin-1 Promote Reendothelialization After Carotid Endarterium Mechanical Injury. Stem Cell Rev Rep 2021; 18:1041-1053. [PMID: 33982245 DOI: 10.1007/s12015-021-10180-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Stanniocalcin-1 (STC-1) is a secreted glycoprotein that participates in the regulation of inflammation, apoptosis, and necrosis. We investigated the reendothelialization effect of exosomes from adipose stem cells (ADSC) overexpressing STC-1 on injured carotid endarterium. METHODS ADSCs were transfected with lentivirus vectors containing pre-STC-1. PHK-26 as molecular probe was used to track the exosomes engulfed by mice arterial endothelial cells (MAEC). The role of STC-1-ADSC-Exosome (S-ADSC-Exo) in MAECs was verified through scratch test and tube forming. Expressions of STC-1 and NLRP3 inflammasome were detected by western blot and quantitative reverse transcription polymerase chain reaction. Reendothelialization effect was inhibited by the antagonist of siRNA targeting STC-1. Carotid endarterium mechanical injury was induced by insertion with a guidewire into the common carotid artery lumen. Carotid arteries were harvested for histological examination, immunofluorescence staining, and Evan's blue staining. RESULTS Transfection of STC-1 significantly enhanced STC-1 levels in ADSCs, their exosomes, and MAECs. Compared with the control group and the ADSC-Exo group, STC-1 enriched exosomes markedly inhibited the expressions of NLRP3, Caspase-1, and IL-1β in MAECs, exhibited good lateral migration capacity, and promoted angiogenesis. Administration of siRNA targeting STC-1 completely abolished down-regulation of NLRP3, Caspase-1, and IL-1β by STC-1 and inhibited effects of S-ADSC-Exo on lateral migration and angiogenesis. In vivo administration of S-ADSC-Exo had reendothelialization effect on post-injury carotid endarterium as evidenced by thinner arterial wall, low-expressed NLRP3 inflammasome, and more living endothelial cells. CONCLUSIONS The reendothelialization effect of exosomes from ADSCs on post-injury carotid endarterium could be enhanced by genetic modification of the exosomes to contain elevated STC-1, possibly through suppression of NLRP3 inflammasome-mediated inflammation.
Collapse
|
133
|
Wang Y, Mo Y, Peng M, Zhang S, Gong Z, Yan Q, Tang Y, He Y, Liao Q, Li X, Wu X, Xiang B, Zhou M, Li Y, Li G, Li X, Zeng Z, Guo C, Xiong W. The influence of circular RNAs on autophagy and disease progression. Autophagy 2021; 18:240-253. [PMID: 33904341 DOI: 10.1080/15548627.2021.1917131] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Circular RNAs (circRNAs) are non-coding RNAs that have attracted considerable attention in recent years. Owing to their distinct circular structure, circRNAs are stable in cells. Autophagy is a catabolic process that helps in the degradation and recycling of harmful or inessential biological macromolecules in cells and enables cells to adapt to stress and changes in the internal and external environments. Evidence has shown that circRNAs influence the course of a disease by regulating autophagy, which indicates that autophagy is involved in the onset and development of various diseases and can affect drug resistance (for example, it affects cisplatin resistance in tumors). In this review, we summarized the role of circRNAs in autophagy and their influence on disease onset and progression as well as drug resistance. The review will expand our understanding of tumors as well as cardiovascular and neurological diseases and also suggest novel therapeutic strategies.Abbreviations: ACR: autophagy-related circRNA; ADSCs: adipogenic mesenchymal stem cells; AMPK: AMP-activated protein kinase; ATG: autophagy related; BCL2: BCL2 apoptosis regulator; BECN1: beclin 1; ceRNA: competing endogenous RNA; circRNA: circular RNA; CMA: chaperone-mediated autophagy; EPCs: endothelial progenitor cells; LE/MVBs: late endosomes/multivesicular bodies; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NSCLC: non-small cell lung cancer; PDLSCs: periodontal ligament stem cells; PE: phosphatidylethanolamine; PtdIns: phosphatidylinositol; PtdIns3K: phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate 1,2-dipalmitoyl; PTEN: phosphatase and tensin homolog; RBPs: RNA-binding proteins; SiO2: silicon dioxide; TFEB: transcription factor EB; ULK: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Yian Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Miao Peng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanyan Tang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi He
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xu Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Bo Xiang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| |
Collapse
|
134
|
Bailey AJM, Li H, Kirkham AM, Tieu A, Maganti HB, Shorr R, Fergusson DA, Lalu MM, Elomazzen H, Allan DS. MSC-Derived Extracellular Vesicles to Heal Diabetic Wounds: a Systematic Review and Meta-Analysis of Preclinical Animal Studies. Stem Cell Rev Rep 2021; 18:968-979. [PMID: 33893619 PMCID: PMC8064883 DOI: 10.1007/s12015-021-10164-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 12/15/2022]
Abstract
Introduction Extracellular vesicles from mesenchymal stromal cells (MSC-EVs) have shown promise in wound healing. Their use in diabetic wounds specifically, however, remains pre-clinical and their efficacy remains uncertain less clear. A systematic review of preclinical studies is needed to determine the efficacy of MSC-EVs in the treatment of diabetic wounds to accelerate the clinical translation of this cell-based therapy. Methods PubMed and Embase were searched (to June 23, 2020). All English-language, full-text, controlled interventional studies comparing MSC-EVs to placebo or a “no treatment” arm in animal models of diabetic wounds were included. Study outcomes, including wound closure (primary outcome), scar width, blood vessel number and density, and re-epithelialisation were pooled using a random effects meta-analysis. Risk of bias (ROB) was assessed using the SYRCLE tool for pre-clinical animal studies. Results A total of 313 unique records were identified from our search, with 10 full text articles satisfying inclusion criteria (n = 136 animals). The administration of MSC-EVs improved closure of diabetic wounds compared to controls with a large observed effect (Standardized Mean Difference (SMD) 5.48, 95% Confidence Interval (CI) 3.55–8.13). Healing was further enhanced using MSC-EVs enriched in non-coding RNAs or microRNAs compared to controls (SMD 9.89, 95%CI 7.32–12.46). Other outcomes, such as blood vessel density and number, scar width, and re-epithelialisation were improved with the administration of MSC-EVs, with a large effect. ROB across studies was unclear. Conclusion MSC-EVs, particularly following enrichment for specific RNAs, are a promising treatment for diabetic wounds in pre-clinical studies and translation to the clinical domain appears warranted. Registration PROSPERO #CRD42020199327 [248]. Graphical abstract Forest plot demonstrating increased wound closure rates of diabetic wounds receiving genetically modified MSC-EVs that were enriched for specific RNAs. DFO = deferoxamine. Control groups were inactive (no treatment or saline) except for 3 studies which used hydrogels without MSC-EVs as control (Li M 2016; Shi 2017; Tao 2016). ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s12015-021-10164-4.
Collapse
Affiliation(s)
- Adrian J M Bailey
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Stem Cells and Centre for Innovation, Canadian Blood Services, Ottawa, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Heidi Li
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Aidan M Kirkham
- Stem Cells and Centre for Innovation, Canadian Blood Services, Ottawa, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Alvin Tieu
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
- Regenerative Medicine Programs, Ottawa Hospital Research Institute, Ottawa, Canada
- Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Harinad B Maganti
- Stem Cells and Centre for Innovation, Canadian Blood Services, Ottawa, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Risa Shorr
- Library and Information Services, The Ottawa Hospital, Ottawa, Canada
| | - Dean A Fergusson
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
- Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Canada
- School of Public Health and Epidemiology, University of Ottawa, Ottawa, Canada
| | - Manoj M Lalu
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
- Regenerative Medicine Programs, Ottawa Hospital Research Institute, Ottawa, Canada
- Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Departments of Anesthesiology and Pain Medicine, University of Ottawa, Ottawa, Canada
| | - Heidi Elomazzen
- Stem Cells and Centre for Innovation, Canadian Blood Services, Ottawa, Canada
| | - David S Allan
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Stem Cells and Centre for Innovation, Canadian Blood Services, Ottawa, Canada.
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada.
- Regenerative Medicine Programs, Ottawa Hospital Research Institute, Ottawa, Canada.
- Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Canada.
- Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
135
|
Geng K, Ma X, Jiang Z, Huang W, Gao C, Pu Y, Luo L, Xu Y, Xu Y. Innate Immunity in Diabetic Wound Healing: Focus on the Mastermind Hidden in Chronic Inflammatory. Front Pharmacol 2021; 12:653940. [PMID: 33967796 PMCID: PMC8097165 DOI: 10.3389/fphar.2021.653940] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
A growing body of evidence suggests that the interaction between immune and metabolic responses is essential for maintaining tissue and organ homeostasis. These interacting disorders contribute to the development of chronic diseases associated with immune-aging such as diabetes, obesity, atherosclerosis, and nonalcoholic fatty liver disease. In Diabetic wound (DW), innate immune cells respond to the Pathogen-associated molecular patterns (PAMAs) and/or Damage-associated molecular patterns (DAMPs), changes from resting to an active phenotype, and play an important role in the triggering and maintenance of inflammation. Furthermore, the abnormal activation of innate immune pathways secondary to immune-aging also plays a key role in DW healing. Here, we review studies of innate immune cellular molecular events that identify metabolic disorders in the local microenvironment of DW and provide a historical perspective. At the same time, we describe some of the recent progress, such as TLR receptor-mediated intracellular signaling pathways that lead to the activation of NF-κB and the production of various pro-inflammatory mediators, NLRP3 inflammatory via pyroptosis, induction of IL-1β and IL-18, cGAS-STING responds to mitochondrial injury and endoplasmic reticulum stress, links sensing of metabolic stress to activation of pro-inflammatory cascades. Besides, JAK-STAT is also involved in DW healing by mediating the action of various innate immune effectors. Finally, we discuss the great potential of targeting these innate immune pathways and reprogramming innate immune cell phenotypes in DW therapy.
Collapse
Affiliation(s)
- Kang Geng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Department of Plastic and Burn Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,National Key Clinical Construction Specialty, Luzhou, China
| | - Xiumei Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Zongzhe Jiang
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Chenlin Gao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Yueli Pu
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Lifang Luo
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China
| | - Yong Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
136
|
Differential Therapeutic Effect of Extracellular Vesicles Derived by Bone Marrow and Adipose Mesenchymal Stem Cells on Wound Healing of Diabetic Ulcers and Correlation to Their Cargoes. Int J Mol Sci 2021; 22:ijms22083851. [PMID: 33917759 PMCID: PMC8068154 DOI: 10.3390/ijms22083851] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stem cells isolated from both bone marrow (BMSCs) and adipose tissue (ADSCs) show potential therapeutic effects. These vesicles often show a similar beneficial effect on tissue regeneration, but in some contexts, they exert different biological properties. To date, a comparison of their molecular cargo that could explain the different biological effect is not available. Here, we demonstrated that ADSC-EVs, and not BMSC-EVs, promote wound healing on a murine model of diabetic wounds. Besides a general similarity, the bioinformatic analysis of their protein and miRNA cargo highlighted important differences between these two types of EVs. Molecules present exclusively in ADSC-EVs were highly correlated to angiogenesis, whereas those expressed in BMSC-EVs were preferentially involved in cellular proliferation. Finally, in vitro analysis confirmed that both ADSC and BMSC-EVs exploited beneficial effect on cells involved in skin wound healing such as fibroblasts, keratinocytes and endothelial cells, but through different cellular processes. Consistent with the bioinformatic analyses, BMSC-EVs were shown to mainly promote proliferation, whereas ADSC-EVs demonstrated a major effect on angiogenesis. Taken together, these results provide deeper comparative information on the cargo of ADSC-EVs and BMSC-EVs and the impact on regenerative processes essential for diabetic wound healing.
Collapse
|
137
|
Quiñones-Vico MI, Sanabria-de la Torre R, Sánchez-Díaz M, Sierra-Sánchez Á, Montero-Vílchez T, Fernández-González A, Arias-Santiago S. The Role of Exosomes Derived From Mesenchymal Stromal Cells in Dermatology. Front Cell Dev Biol 2021; 9:647012. [PMID: 33898436 PMCID: PMC8058372 DOI: 10.3389/fcell.2021.647012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
The skin is the largest organ of the human body and its main functions include providing protection from external harmful agents, regulating body temperature, and homeostatic maintenance. Skin injuries can damage this important barrier and its functions so research focuses on approaches to accelerate wound healing and treat inflammatory skin diseases. Due to their regenerative and immunomodulatory properties, mesenchymal stromal cells (MSCs) have been reported to play a significant role in skin repair and regeneration. However, it seems that the secretome of these cells and exosomes in particular may be responsible for their functions in skin regeneration and the immunomodulation field. The present review aims to gather the available information about the role of MSC-derived exosomes for both in vitro and in vivo models of different skin conditions and to highlight the need for further research in order to overcome any limitations for clinical translation.
Collapse
Affiliation(s)
- María I. Quiñones-Vico
- Cell Production and Tissue Engineering Unit, Virgen de las Nieves University Hospital, Granada, Spain
- Granada Biosanitary Research Institute (ibs. GRANADA), Granada, Spain
- Andalusian Network for the Design and Translation of Advanced Therapies, Seville, Spain
- Department of Dermatology, School of Medicine, University of Granada, Granada, Spain
| | - Raquel Sanabria-de la Torre
- Cell Production and Tissue Engineering Unit, Virgen de las Nieves University Hospital, Granada, Spain
- Granada Biosanitary Research Institute (ibs. GRANADA), Granada, Spain
- Andalusian Network for the Design and Translation of Advanced Therapies, Seville, Spain
| | - Manuel Sánchez-Díaz
- Granada Biosanitary Research Institute (ibs. GRANADA), Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Álvaro Sierra-Sánchez
- Cell Production and Tissue Engineering Unit, Virgen de las Nieves University Hospital, Granada, Spain
- Granada Biosanitary Research Institute (ibs. GRANADA), Granada, Spain
- Andalusian Network for the Design and Translation of Advanced Therapies, Seville, Spain
| | - Trinidad Montero-Vílchez
- Granada Biosanitary Research Institute (ibs. GRANADA), Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Ana Fernández-González
- Cell Production and Tissue Engineering Unit, Virgen de las Nieves University Hospital, Granada, Spain
- Granada Biosanitary Research Institute (ibs. GRANADA), Granada, Spain
- Andalusian Network for the Design and Translation of Advanced Therapies, Seville, Spain
| | - Salvador Arias-Santiago
- Cell Production and Tissue Engineering Unit, Virgen de las Nieves University Hospital, Granada, Spain
- Granada Biosanitary Research Institute (ibs. GRANADA), Granada, Spain
- Andalusian Network for the Design and Translation of Advanced Therapies, Seville, Spain
- Department of Dermatology, School of Medicine, University of Granada, Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, Granada, Spain
| |
Collapse
|
138
|
Li J, Wei M, Liu X, Xiao S, Cai Y, Li F, Tian J, Qi F, Xu G, Deng C. The progress, prospects, and challenges of the use of non-coding RNA for diabetic wounds. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:554-578. [PMID: 33981479 PMCID: PMC8063712 DOI: 10.1016/j.omtn.2021.03.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic diabetic wounds affect the quality of life of patients, resulting in significant social and economic burdens on both individuals and the health care system. Although treatment methods for chronic diabetic wounds have been explored, there remains a lack of effective treatment strategies; therefore, alternative strategies must be explored. Recently, the abnormal expression of non-coding RNA in diabetic wounds has received widespread attention since it is an important factor in the development of diabetic wounds. This article reviews the regulatory role of three common non-coding RNAs (microRNA [miRNA], long non-coding RNA [lncRNA], and circular RNA [circRNA]) in diabetic wounds and discusses the diagnosis, treatment potential, and challenges of non-coding RNA in diabetic wounds. This article provides insights into new strategies for diabetic wound diagnosis and treatment at the genetic and molecular levels.
Collapse
Affiliation(s)
- Jianyi Li
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Miaomiao Wei
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Xin Liu
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Shune Xiao
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Yuan Cai
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Fang Li
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Jiao Tian
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Fang Qi
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Guangchao Xu
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Chengliang Deng
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China.,Collaborative Innovation Center of Tissue Injury Repair and Regenerative Medicine Co-sponsored by Province and Ministry, Affiliated Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| |
Collapse
|
139
|
An T, Chen Y, Tu Y, Lin P. Mesenchymal Stromal Cell-Derived Extracellular Vesicles in the Treatment of Diabetic Foot Ulcers: Application and Challenges. Stem Cell Rev Rep 2021; 17:369-378. [PMID: 32772239 DOI: 10.1007/s12015-020-10014-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetic foot ischemia and ulcer (DFU) persists as a serious diabetes mellitus complication in spite of increased understanding of the pathophysiology and the cellular and molecular responses. Contributing to this pessimistic situation is the lack of effective treatments that are slow to heal the deep chronic wounds and microvascular obstruction. Mesenchymal stromal cells (MSCs) have been tested as a promising cell-based therapy for diabetes in vitro and in vivo, which is able to accelerate wound closure with increased epithelialization, granulation tissue formation and angiogenesis by differentiation into skin cells and paracrine pathways to repair injured cells. The secretomes of MSCs, including cytokines, growth factors, chemokines, and extracellular vesicles containing mRNA, proteins and microRNAs, have immunomodulatory and regenerative effects. This review will shed new light on the therapeutic potential of MSC-derived extracellular vesicles (MSC-EVs) for the treatment of diabetes-induced lower limb ischemia and ulcers. The identification of underlying mechanisms for MSC-EVs regulation on impaired diabetic wound healing might provide a new direction for MSC-centered treatment for diabetic lower limb ischemia and ulcers. Immunomodulatory and angiogenic effects of MSC-derived extracellular vesicles on diabetic foot ulcer.
Collapse
Affiliation(s)
- Tao An
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China
| | - Yi Chen
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China
| | - Yingchun Tu
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China
| | - Ping Lin
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China.
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China.
| |
Collapse
|
140
|
Xiang Y, Kuai L, Ru Y, Jiang J, Li X, Li F, Chen Q, Li B. Transcriptional profiling and circRNA-miRNA-mRNA network analysis identify the biomarkers in Sheng-ji Hua-yu formula treated diabetic wound healing. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113643. [PMID: 33271241 DOI: 10.1016/j.jep.2020.113643] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/14/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sheng-ji Hua-yu (SJHY) formula is a traditional Chinese herbal which is effective in treating diabetic ulcers. It has been indicated to accelerate re-epithelialization and healing time of cutaneous wounds in a Streptozotocin (STZ)-induced diabetic mouse model. However, its mechanisms remain undetermined. AIM OF THE STUDY To reveal the molecular mechanisms of SJHY formula in treating diabetic wounds through transcriptional profiling and circRNA-miRNA-mRNA network analysis clues. MATERIALS AND METHODS Protein expressions of tumor necrosis factor (TNF-α), interleukin (IL)-6, IL-1β in skin tissues of wounds from SJHY formula-treated and untreated mice were analyzed by Bio-Plex assay. Differentially expressed (DE) genes were detected by whole transcriptome sequencing (RNA-seq). Using predicted miRNA targets, circRNA-miRNA-mRNA networks were constructed. Furthermore, quantitative real-time PCR (qRT-PCR) was utilized to validate the circRNA-miRNA-mRNA networks. RESULTS Bio-Plex assay illustrated that the protein expressions of TNF-α, IL-1β, IL-6 were downregulated in SJHY formula-treated diabetic wounds compared with untreated wounds. RNA-seq identified 11 DE circRNAs and 476 DE mRNAs between SJHY formula-treated and diabetic mice, including 4 upregulated and 7 downregulated circRNAs, 311 upregulated and 165 downregulated mRNAs. CircRNA-Krt13/miR-665-3p/Itga3 and circRNA-Krt14/miR-706/Mylk4 pathways were built, which may contribute to the healing of SJHY formula-treated diabetic wounds. CONCLUSIONS Overall, this study suggests that these 2 circRNA-miRNA-mRNA networks are potential biomarkers for evaluation of SJHY formula efficacy in diabetic wound healing, which provides evidence to support its clinical applications.
Collapse
Affiliation(s)
- Yanwei Xiang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi Ru
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jingsi Jiang
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Fulun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qilong Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
141
|
van de Vyver M, Powrie YSL, Smith C. Targeting Stem Cells in Chronic Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1286:163-181. [PMID: 33725353 DOI: 10.1007/978-3-030-55035-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cell (MSC) dysfunction is a serious complication in ageing and age-related inflammatory diseases such as type 2 diabetes mellitus. Inflammation and oxidative stress-induced cellular senescence alter the immunomodulatory ability of MSCs and hamper their pro-regenerative function, which in turn leads to an increase in disease severity, maladaptive tissue damage and the development of comorbidities. Targeting stem/progenitor cells to restore their function and/or protect them against impairment could thus improve healing outcomes and significantly enhance the quality of life for diabetic patients. This review discusses the dysregulation of MSCs' immunomodulatory capacity in the context of diabetes mellitus and focuses on intervention strategies aimed at MSC rejuvenation. Research pertaining to the potential therapeutic use of either pharmacological agents (NFкB antagonists), natural products (phytomedicine) or biological agents (exosomes, probiotics) to improve MSC function is discussed and an overview of the most pertinent methodological considerations given. Based on in vitro studies, numerous anti-inflammatory agents, antioxidants and biological agents show tremendous potential to revitalise MSCs. An integrated systems approach and a thorough understanding of complete disease pathology are however required to identify feasible candidates for in vivo targeting of MSCs.
Collapse
Affiliation(s)
- Mari van de Vyver
- Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, Cape Town, South Africa.
| | - Yigael S L Powrie
- Department of Medicine, Faculty of Medicine & Health Sciences, Stellenbosch University, Cape Town, South Africa.,Department of Physiological Sciences, Science Faculty, Stellenbosch University, Stellenbosch, South Africa
| | - Carine Smith
- Department of Physiological Sciences, Science Faculty, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
142
|
Dalirfardouei R, Gholoobi A, Vahabian M, Mahdipour E, Afzaljavan F. Therapeutic role of extracellular vesicles derived from stem cells in cutaneous wound models: A systematic review. Life Sci 2021; 273:119271. [PMID: 33652035 DOI: 10.1016/j.lfs.2021.119271] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/07/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence has shown that extracellular vesicles can be efficient as experimental therapeutics in pre-clinical models of skin wounds, but there is a significant unmet need to translate this to clinical utilization. The objectives of the current systematic review were to identify the strength of the therapeutic effects of EVs derived from stem cells in cutaneous wounds and to assess which EV-mediated mechanisms could be involved in the therapeutic response. PubMed, ISI Web of Science, and Scopus databases were systematically searched. We retrieved English-language articles published through June 2020. In vivo studies which applied stem cell-derived EVs were included for further analysis. The Risk of bias was assessed by the SYRCLE tool. We identified thirty-nine pre-clinical studies that evaluated the effects of EVs on the wound healing process. The included studies varied greatly in EVs isolation techniques, route of administration, EVs producing cells, and follow-up time. In vivo application revealed beneficial effects of EVs on accelerating wound closure and re-epithelialization in a dose-dependent manner. Elevated angiogenesis was reported in twelve eligible studies through multiple signaling pathways such as PI3K/Akt, MAPK/ERK, and JAK/STAT. The well-known signaling pathway to inhibit scar formation was TGF-β2/SMAD2. However, all included studies were not blinded enough which may have introduced bias. Therefore, the transition of EV's efficacy into the clinics is deeply rooted in the following important factors: 1) pre-clinical studies with a lower risk of bias and longer follow-up time, and 2) consistent, reproducible, and feasible manufacturing of EVs production in a large-scale commercial program.
Collapse
Affiliation(s)
- Razieh Dalirfardouei
- Research center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Aida Gholoobi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrangiz Vahabian
- Department of English Language and Persian Literature, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elahe Mahdipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
143
|
Gurunathan S, Kang MH, Kim JH. A Comprehensive Review on Factors Influences Biogenesis, Functions, Therapeutic and Clinical Implications of Exosomes. Int J Nanomedicine 2021; 16:1281-1312. [PMID: 33628021 PMCID: PMC7898217 DOI: 10.2147/ijn.s291956] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/16/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nanoscale-sized membrane vesicles secreted by almost all cell types into the extracellular environment upon fusion of multivesicular bodies and plasma membrane. Biogenesis of exosomes is a protein quality control mechanism, and once released, exosomes transmit signals to other cells. The applications of exosomes have increased immensely in biomedical fields owing to their cell-specific cargos that facilitate intercellular communications with neighboring cells through the transfer of biologically active compounds. The diverse constituents of exosomes reflect their cell of origin and their detection in biological fluids represents a diagnostic marker for various diseases. Exosome research is expanding rapidly due to the potential for clinical application to therapeutics and diagnosis. However, several aspects of exosome biology remain elusive. To discover the use of exosomes in the biomedical applications, we must better understand the basic molecular mechanisms underlying their biogenesis and function. In this comprehensive review, we describe factors involved in exosomes biogenesis and the role of exosomes in intercellular signaling and cell-cell communications, immune responses, cellular homeostasis, autophagy, and infectious diseases. In addition, we discuss the role of exosomes as diagnostic markers, and their therapeutic and clinical implications. Furthermore, we addressed the challenges and outstanding developments in exosome research, and discuss future perspectives.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| |
Collapse
|
144
|
Xiong M, Zhang Q, Hu W, Zhao C, Lv W, Yi Y, Wang Y, Tang H, Wu M, Wu Y. The novel mechanisms and applications of exosomes in dermatology and cutaneous medical aesthetics. Pharmacol Res 2021; 166:105490. [PMID: 33582246 DOI: 10.1016/j.phrs.2021.105490] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/18/2021] [Accepted: 02/09/2021] [Indexed: 12/17/2022]
Abstract
Exposure to the external environment may lead to instability and dysfunction of the skin, resulting in refractory wound, skin aging, pigmented dermatosis, hair loss, some immune-mediated dermatoses, and connective tissue diseases. Nowadays, many skin treatments have not achieved a commendable balance between medical recovery and cosmetic needs. Exosomes are cell-derived nanoscale vesicles carrying various biomolecules, including proteins, nucleic acids, and lipids, with the capability to communicate with adjacent or distant cells. Recent studies have demonstrated that endogenic multiple kinds of exosomes are crucial orchestrators in shaping physiological and pathological development of the skin. Besides, exogenous exosomes, such as stem cell exosomes, can serve as novel treatment options to repair, regenerate, and rejuvenate skin tissue. Herein, we review new insights into the role of endogenic and exogenous exosomes in the skin microenvironment and recent advances in applications of exosomes related to dermatology and cutaneous medical aesthetics. The deep understanding of the mechanisms by which exosomes perform biological functions in skin is of great potential to establish attractive therapeutic methods for the skin.
Collapse
Affiliation(s)
- Mingchen Xiong
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China.
| | - Weijie Hu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China.
| | - Chongru Zhao
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Wenchang Lv
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Yi Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Yichen Wang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Hongbo Tang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China.
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China.
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China.
| |
Collapse
|
145
|
Xiong J, Hu H, Guo R, Wang H, Jiang H. Mesenchymal Stem Cell Exosomes as a New Strategy for the Treatment of Diabetes Complications. Front Endocrinol (Lausanne) 2021; 12:646233. [PMID: 33995278 PMCID: PMC8117220 DOI: 10.3389/fendo.2021.646233] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease, now prevalent worldwide, which is characterized by a relative or absolute lack of insulin secretion leading to chronically increased blood glucose levels. Diabetic patients are often accompanied by multiple macrovascular complications, such as coronary heart disease, hypertension, macrovascular arteriosclerosis, and microvascular complications. Microvascular complications include diabetic kidney injury, diabetic encephalopathy, and diabetic foot, which reduce the quality of life and survival status of patients. Mesenchymal stem cell exosomes (MSC-Exos) possess repair functions similar to MSCs, low immunogenicity, and ease of storage and transport. MSC-Exos have been proven to possess excellent repair effects in repairing various organ damages. This study reviews the application of MSC-Exos in the treatment of DM and its common complications. MSC-Exos may be used as an effective treatment for DM and its complications.
Collapse
Affiliation(s)
| | | | | | - Hui Wang
- *Correspondence: Hui Wang, ; Hua Jiang,
| | - Hua Jiang
- *Correspondence: Hui Wang, ; Hua Jiang,
| |
Collapse
|
146
|
Li FXZ, Lin X, Xu F, Shan SK, Guo B, Lei LM, Zheng MH, Wang Y, Xu QS, Yuan LQ. The Role of Mesenchymal Stromal Cells-Derived Small Extracellular Vesicles in Diabetes and Its Chronic Complications. Front Endocrinol (Lausanne) 2021; 12:780974. [PMID: 34987478 PMCID: PMC8721875 DOI: 10.3389/fendo.2021.780974] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are applied in regenerative medicine of several tissues and organs nowadays by virtue of their self-renewal capabilities, multiple differentiation capacity, potent immunomodulatory properties, and their ability to be favourably cultured and manipulated. With the continuous development of "cell-free therapy" research, MSC-derived small extracellular vesicles (MSC-sEVs) have increasingly become a research hotspot in the treatment of various diseases. Small extracellular vesicles (SEVs) are membrane vesicles with diameters of 30 to 150 nm that mediate signal transduction between adjacent or distal cells or organs by delivering non-coding RNA, protein, and DNA. The contents and effects of sEVs vary depending on the properties of the originating cell. In recent years, MSC-sEVs have been found to play an important role in the occurrence and development of diabetes mellitus as a new way of communication between cells. Diabetes mellitus is a common metabolic disease in clinic. Its complications of the heart, brain, kidney, eyes, and peripheral nerves are a serious threat to human health and has been a hot issue for clinicians. MSC-sEVs could be applied to repair or prevent damage from the complications of diabetes mellitus through anti-inflammatory effects, reduction of endoplasmic reticulum-related protein stress, polarization of M2 macrophages, and increasing autophagy. Therefore, we highly recommend that MSC-sEVs-based therapies to treat diabetes mellitus and its chronic complication be further explored. The analysis of the role and molecular mechanisms of MSC-sEVs in diabetes and its related complications will provide new idea and insights for the prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Fu-Xing-Zi Li
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Wang
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- National Clinical Research Center for Metabolic Disease, Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Ling-Qing Yuan,
| |
Collapse
|
147
|
Vu NB, Nguyen HT, Palumbo R, Pellicano R, Fagoonee S, Pham PV. Stem cell-derived exosomes for wound healing: current status and promising directions. Minerva Med 2020; 112:384-400. [PMID: 33263376 DOI: 10.23736/s0026-4806.20.07205-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Wound healing, especially of chronic wounds, is still an unmet therapeutic area since assessment and management are extremely complicated. Although many efforts have been made to treat wounds, all strategies have achieved limited results for chronic wounds. Stem cell-based therapy is considered a promising approach for complex wounds such as those occurring in diabetics. Mesenchymal stem cell transplantation significantly improves wound closure, angiogenesis and wound healing. However, cell therapy is complex, expensive and time-consuming. Recent studies have shown that stem cell-derived exosomes can be an exciting approach to treat wounds. Exosomes derived from mesenchymal stem cells can induce benefit in almost all stages of wound healing, including control of immune responses, inhibition of inflammation, promoting cell proliferation and angiogenesis, while reducing scar formation during the wound healing process. This review aimed at offering an updated overview of the use of exosomes in biological applications, such as wound healing, and addresses not only current applications but also new directions for this next-generation approach in wound healing.
Collapse
Affiliation(s)
- Ngoc B Vu
- Stem Cell Institute, University of Science, Ho Chi Minh, Vietnam.,Vietnam National University - Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Hoa T Nguyen
- Stem Cell Institute, University of Science, Ho Chi Minh, Vietnam.,Vietnam National University - Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Rosanna Palumbo
- Institute of Biostructure and Bioimaging (CNR), Naples, Italy
| | | | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| | - Phuc V Pham
- Stem Cell Institute, University of Science, Ho Chi Minh, Vietnam - .,Vietnam National University - Ho Chi Minh City, Ho Chi Minh, Vietnam.,Laboratory of Stem Cell Research and Application, Ho Chi Minh, Vietnam
| |
Collapse
|
148
|
Shang Q, Shen G, Chen G, Zhang Z, Yu X, Zhao W, Zhang P, Chen H, Tang K, Yu F, Tang J, Liang D, Jiang X, Ren H. The emerging role of miR-128 in musculoskeletal diseases. J Cell Physiol 2020; 236:4231-4243. [PMID: 33241566 DOI: 10.1002/jcp.30179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/24/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022]
Abstract
MicroRNA-128 (miR-128) is associated with cell proliferation, differentiation, migration, apoptosis, and survival. Genetic analysis studies have demonstrated that miR-128 participates in bone metabolism, which involves bone marrow-derived mesenchymal stem cells, osteoblasts, osteoclasts, and adipocytes. miR-128 also participates in regeneration of skeletal muscles by targeting myoblast-associated proteins. The deregulation of miR-128 could lead to a series of musculoskeletal diseases. In this review, we discuss recent findings of miR-128 in relation to bone metabolism and muscle regeneration to determine its potential therapeutic effects in musculoskeletal diseases, and to propose directions for future research in this significant field.
Collapse
Affiliation(s)
- Qi Shang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengyang Shen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guifeng Chen
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhua Zhao
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Zhang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglin Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kai Tang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fuyong Yu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Tang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaobing Jiang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Ren
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
149
|
Petkovic M, Sørensen AE, Leal EC, Carvalho E, Dalgaard LT. Mechanistic Actions of microRNAs in Diabetic Wound Healing. Cells 2020; 9:E2228. [PMID: 33023156 PMCID: PMC7601058 DOI: 10.3390/cells9102228] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Wound healing is a complex biological process that is impaired under diabetes conditions. Chronic non-healing wounds in diabetes are some of the most expensive healthcare expenditures worldwide. Early diagnosis and efficacious treatment strategies are needed. microRNAs (miRNAs), a class of 18-25 nucleotide long RNAs, are important regulatory molecules involved in gene expression regulation and in the repression of translation, controlling protein expression in health and disease. Recently, miRNAs have emerged as critical players in impaired wound healing and could be targets for potential therapies for non-healing wounds. Here, we review and discuss the mechanistic background of miRNA actions in chronic wounds that can shed the light on their utilization as specific wound healing biomarkers.
Collapse
Affiliation(s)
- Marija Petkovic
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark; (A.E.S.); (L.T.D.)
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (E.C.L.); (E.C.)
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Anja Elaine Sørensen
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark; (A.E.S.); (L.T.D.)
| | - Ermelindo Carreira Leal
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (E.C.L.); (E.C.)
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Eugenia Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (E.C.L.); (E.C.)
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Department of Geriatrics, University of Arkansas for Medical Sciences, and Arkansas Children’s Research Institute, Little Rock, AR 72205, USA
| | - Louise Torp Dalgaard
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark; (A.E.S.); (L.T.D.)
| |
Collapse
|
150
|
Xiong M, Zhang Q, Hu W, Zhao C, Lv W, Yi Y, Wu Y, Wu M. Exosomes From Adipose-Derived Stem Cells: The Emerging Roles and Applications in Tissue Regeneration of Plastic and Cosmetic Surgery. Front Cell Dev Biol 2020; 8:574223. [PMID: 33015067 PMCID: PMC7511773 DOI: 10.3389/fcell.2020.574223] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
Adipose-derived stem cells (ASCs) are an important stem cell type separated from adipose tissue, with the properties of multilineage differentiation, easy availability, high proliferation potential, and self-renewal. Exosomes are novel frontiers of intercellular communication regulating the biological behaviors of cells, such as angiogenesis, immune modulation, proliferation, and migration. ASC-derived exosomes (ASC-exos) are important components released by ASCs paracrine, possessing multiple biological activities. Tissue regeneration requires coordinated “vital networks” of multiple growth factors, proteases, progenitors, and immune cells producing inflammatory cytokines. Recently, as cell-to-cell messengers, ASC-exos have received much attention for the fact that they are important paracrine mediators contributing to their suitability for tissue regeneration. ASC-exos, with distinct properties by encapsulating various types of bioactive cargoes, are endowed with great application potential in tissue regeneration, mechanically via the migration and proliferation of repair cells, facilitation of the neovascularization, and other specific functions in different tissues. Here, this article elucidated the research progress of ASC-exos about tissue regeneration in plastic and cosmetic surgery, including skin anti-aging therapy, dermatitis improvement, wound healing, scar removal, flap transplantation, bone tissue repair and regeneration, obesity prevention, fat grafting, breast cancer, and breast reconstruction. Deciphering the biological properties of ASC-exos will provide further insights for exploring novel therapeutic strategies of tissue regeneration in plastic and cosmetic surgery.
Collapse
Affiliation(s)
- Mingchen Xiong
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijie Hu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chongru Zhao
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenchang Lv
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|