1
|
Yang D, Yang J, Chang SJ, Hu JL, Chen YJ, Yang SW. Exosome-Seeded Cryogel Scaffolds for Extracellular Matrix Regeneration in the Repair of Articular Cartilage Defects: An In Vitro and In Vivo Rabbit Model Study. Polymers (Basel) 2025; 17:975. [PMID: 40219364 PMCID: PMC11991529 DOI: 10.3390/polym17070975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Traumatic or degenerative defects of articular cartilage impair joint function, and the treatment of articular cartilage damage remains a challenge. By mimicking the cartilage extracellular matrix (ECM), exosome-seeded cryogels may enhance cell proliferation and chondral repair. ECM-based cryogels were cryopolymerized with gelatin, chondroitin sulfate, and various concentrations (0%, 0.3%, 0.5%, and 1%) of hyaluronic acid (HA), and their water content, swelling ratio, porosity, mechanical properties, and effects on cell viability were evaluated. The regenerative effects of bone marrow-derived mesenchymal stem cell (BM-MSC)-derived exosome (at a concentration of 106 particles/mL)-seeded 0.3% HA cryogels were assessed in vitro and in surgically induced male New Zealand rabbit cartilage defects in vivo. The water content, swelling ratio, and porosity of the cryogels significantly (p < 0.05) increased and the Young's modulus values of the cryogels decreased with increasing HA concentrations. MTT assays revealed that the developed biomaterials had no cytotoxic effects. The optimal cryogel composition was 0.3% HA, and the resulting cryogel had favorable properties and suitable mechanical strength. Exosomes alone and exosome-seeded cryogels promoted chondrocyte proliferation (with cell optical densities that were 58% and 51% greater than that of the control). The cryogel alone and the exosome-seeded cryogel facilitated ECM deposition and sulfated glycosaminoglycan synthesis. Although we observed cartilage repair via Alcian blue staining with both the cryogel alone and the exosome-seeded cryogel, the layered arrangement of the chondrocytes was superior to that of the control chondrocytes when exosome-seeded cryogels were used. This study revealed the potential value of using BM-MSC-derived exosome-seeded ECM-based cryogels for cartilage tissue engineering to treat cartilage injury.
Collapse
Affiliation(s)
- Daniel Yang
- Laboratory of Regenerative Medicine and Biosensors, I-Shou University, Kaohsiung City 824005, Taiwan; (D.Y.); (J.Y.); (S.-J.C.); (J.-L.H.); (Y.-J.C.)
- Cambridge International Programme, St. Dominic Catholic High School, Kaohsiung City 802306, Taiwan
| | - Joseph Yang
- Laboratory of Regenerative Medicine and Biosensors, I-Shou University, Kaohsiung City 824005, Taiwan; (D.Y.); (J.Y.); (S.-J.C.); (J.-L.H.); (Y.-J.C.)
- Cambridge International Programme, St. Dominic Catholic High School, Kaohsiung City 802306, Taiwan
| | - Shwu-Jen Chang
- Laboratory of Regenerative Medicine and Biosensors, I-Shou University, Kaohsiung City 824005, Taiwan; (D.Y.); (J.Y.); (S.-J.C.); (J.-L.H.); (Y.-J.C.)
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City 824005, Taiwan
| | - Jhe-Lun Hu
- Laboratory of Regenerative Medicine and Biosensors, I-Shou University, Kaohsiung City 824005, Taiwan; (D.Y.); (J.Y.); (S.-J.C.); (J.-L.H.); (Y.-J.C.)
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City 824005, Taiwan
| | - Yong-Ji Chen
- Laboratory of Regenerative Medicine and Biosensors, I-Shou University, Kaohsiung City 824005, Taiwan; (D.Y.); (J.Y.); (S.-J.C.); (J.-L.H.); (Y.-J.C.)
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City 824005, Taiwan
| | - Shan-Wei Yang
- Department of Orthopedics, Kaohsiung Veterans General Hospital, Kaohsiung City 813414, Taiwan
- School of Nursing, Fooyin University, Kaohsiung City 831301, Taiwan
- Department of Leisure and Sports Management, Cheng Shiu University, Kaohsiung City 833301, Taiwan
| |
Collapse
|
2
|
Li Q, Guo R, Zhao C, Chen X, Wang H, Shen C. End Plate Chondrocyte-Derived Exosomal miR-133a-3p Alleviates Intervertebral Disc Degeneration by Targeting the NF-κB Signaling Pathway through the miR-133a-3p/MAML1 Axis. Mol Pharm 2025; 22:1262-1279. [PMID: 39898539 DOI: 10.1021/acs.molpharmaceut.4c00962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Chondrocyte-derived exosomes have shown efficacy in differentiating osteoarthritis-affected cartilage. Intervertebral disc degeneration (IVDD) and osteoarthritis often affect facet joints of the spine and show common epidemiological and pathophysiological characteristics. However, the potential of chondrocyte-derived exosomes for treating IVDD remains unclear. The present study aimed to confirm the effect of end plate chondrocyte-derived exosomes (EPC-Exo) on IVDD and elucidate the underlying mechanism. EPC-Exos were isolated and identified by ultracentrifugation, Western blotting, electron microscopy, and nanoparticle tracking analysis. In the in vitro, EPC-Exo uptake by nucleus pulposus (NP) cells reduced cell death by blocking the nuclear factor-κB (NF-κB) signaling pathway. In the in vivo study, EPC-Exos injected into rat intervertebral discs mitigated lipopolysaccharide-induced IVDD, as revealed by a decreased loss of disc height and improved magnetic resonance imaging findings and histological scores. Bioinformatics and sequencing analyses indicated that EPC-Exos alleviated IVDD through the miR-133a-3p/MAML1 axis. The present study suggests that EPC-Exos reduced IVDD incidence via the miR-133a-3p/MAML1 axis-mediated suppression of NF-κB signaling, which prevented the pyroptosis of NP cells.
Collapse
Affiliation(s)
- Qiuwei Li
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| | - Ruocheng Guo
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| | - Chenhao Zhao
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xuewu Chen
- Spinal Surgery Division of Yijishan Hospital and Wannan Medical College in Wuhu, Wuhu, Anhui 241000, China
| | - Hong Wang
- Spinal Surgery Division of Yijishan Hospital and Wannan Medical College in Wuhu, Wuhu, Anhui 241000, China
| | - Cailiang Shen
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| |
Collapse
|
3
|
Yang G, Ji B, Li H, Liu X, Liu G, Sun J, Yao Y, Li Y, Liu S, Xiao W. Inhibition of CRLF1 expression by miR-8485 alleviates IL-1β-induced chondrocyte inflammation, apoptosis, and extracellular matrix degradation. Int Immunopharmacol 2025; 144:113643. [PMID: 39580860 DOI: 10.1016/j.intimp.2024.113643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 10/22/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
The aim of this study was to investigate the impact of differentially expressed miR-8485 on chondrocyte inflammation in osteoarthritis (OA) and its underlying pathological mechanisms. MiR-8485, which was downregulated in OA, was identified by microarray analysis, and was also found to be decreased in IL-1β-induced C28/I2 cells. miR-8485 down-regulation or IL-1β treated of C28/I2 cells induces a decrease in cellular activity, an increase in apoptosis, an elevation in Cleaved caspase-3, MMP13, and ADAMTS5 protein levels, a decrease in Collagen II and Aggrecan levels, and an increase in the levels of pro-inflammatory factors TNF-α and IL-6. CRLF1 was identified to be a downstream target gene of miR-8485 using bioinformatics prediction and dual luciferase reporter gene assays. CRLF1 was shown to be increased in IL-1β-treated C28/I2 cells, and CRLF1 overexpression partially abrogated the suppressive effect of upregulated miR-8485 on chondrocyte inflammation. In addition, miR-8485 was able to inhibit MAPK/ERK and PI3K/AKT signaling activation by inhibiting CRLF1. In conclusion, miR-8485 was able to inhibit CRLF1 expression and thus inhibit IL-1β-triggered inflammation in chondrocytes, potentially through the inhibition of MAPK/ERK and PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Guang Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Bingzhou Ji
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiu Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Gaoming Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jianfeng Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yuming Yao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shuguang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
4
|
Nikhil A, Gugjoo MB, Das A, Manzoor T, Ahmad SM, Ganai NA, Kumar A. Multilayered Cryogel Enriched with Exosomes Regenerates and Maintains Cartilage Architecture and Phenotype in Goat Osteochondral Injuries. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64505-64521. [PMID: 39555858 DOI: 10.1021/acsami.4c13808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Treatment of critical-size osteochondral (OC) injuries at load-bearing sites has remained a major clinical challenge in orthopedic surgery. This is due to the anisotropic characteristics of OC tissue and the stratified structure of the cartilage. Here, we developed a multilayered OC scaffold by employing cryogelation technology. Gelatin, chitosan, and chondroitin sulfate were utilized for designing three distinct, 2425 ± 120 μm thick layers of cartilage having different alignments, while nanohydroxyapatite and gelatin were used for the subchondral bone layer. Exosomes derived from articular chondrocytes in the range of 60-110 nm were used to promote chondrogenesis. The biocompatibility and cartilage formation potential of the scaffold and exosomes were initially evaluated in rat OC defects. The application of exosome-loaded scaffolds was then investigated in a critical-size OC injury (8 × 10 mm) created in the goat knee. Artificial synovial fluid was designed and utilized as a carrier for exosomes for a booster dose administered as an intra-articular injection. X-ray imaging and micro-CT analysis revealed that the treatment resulted in improved subchondral bone regeneration. The defect region exhibited healthy hyaline cartilage formation, as detected by MRI imaging. Moreover, histological examination revealed that the treatment group showed augmented cell proliferation, matrix deposition, secretion of proteoglycans, and the formation of stratified hyaline cartilage over a long-term (6 and 12 months), whereas the control group demonstrated the formation of fibrocartilage. Treatment-induced upregulation of collagen II, aggrecan, and SOX 9 genes (∼10 fold) further provided evidence that the cartilage phenotype was well preserved. Hence, the proposed treatment has significant translational potential for treating adverse OC clinical injuries.
Collapse
Affiliation(s)
- Aman Nikhil
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Mudasir Bashir Gugjoo
- Division of Veterinary Clinical Complex, SKUAST-Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Ankita Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Tasaduq Manzoor
- Division of Animal Biotechnology, SKUAST-Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology, SKUAST-Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Nazir Ahmad Ganai
- Division of Animal Breeding and Genetics, SKUAST-Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- Centre of Excellence for Materials in Medicine, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
5
|
Han Y, Dong Y, Jia B, Shi X, Zhao H, Li S, Wang H, Sun B, Yin L, Dai K. High-precision bioactive scaffold with dECM and extracellular vesicles targeting 4E-BP inhibition for cartilage injury repair. Mater Today Bio 2024; 27:101114. [PMID: 39211509 PMCID: PMC11360177 DOI: 10.1016/j.mtbio.2024.101114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 09/04/2024] Open
Abstract
The restoration of cartilage injuries remains a formidable challenge in orthopedics, chiefly attributed to the absence of vascularization and innervation in cartilage. Decellularized extracellular matrix (dECM) derived from cartilage, following antigenic removal through decellularization processes, has exhibited remarkable biocompatibility and bioactivity, rendering it a viable candidate for cartilage repair. Additionally, extracellular vesicles (EVs) generated from cartilage have demonstrated a synergistic effect when combined with dECM, potentially mitigating the inhibitory impact on protein synthesis by phosphorylating 4ebp, thereby promoting the synthesis of cartilage-related proteins such as collagen. In pursuit of this objective, we have innovated a novel bioink and repair scaffold characterized by exceptional biocompatibility, bioactivity, and biodegradability, establishing a tissue-specific microenvironment conducive to chondrogenesis. Within rat osteochondral defects, the biologically active scaffold successfully prompted the formation of transparent cartilage, featuring adequate mechanical strength, favorable elasticity, and dECM deposition indicative of cartilage. In summary, this study has effectively engineered a hydrogel bioink tailored for cartilage repair and devised a bioactive cartilage repair scaffold proficient in instigating cell differentiation and fostering cartilage repair.
Collapse
Affiliation(s)
- Yu Han
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Orthopedic Surgery, Shanghai Key Laboratory of Orthopedic Implants,Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yixin Dong
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Bo Jia
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiangyu Shi
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongbo Zhao
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shushan Li
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Haitao Wang
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Binbin Sun
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Li Yin
- Department of Orthopaedics, Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kerong Dai
- Department of Orthopedic Surgery, Shanghai Key Laboratory of Orthopedic Implants,Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
6
|
Lueangarun S, Cho BS, Tempark T. Hair repigmentation of poliosis circumscripta in androgenetic alopecia patient treated with exosomes and fractional picosecond laser. J Cosmet Dermatol 2024; 23:2307-2311. [PMID: 38419400 DOI: 10.1111/jocd.16261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Affiliation(s)
- Suparuj Lueangarun
- Department of Aesthetic Medicine, College of Integrative Medicine, Dhurakij Pundit University, Bangkok, Thailand
- Division of Dermatology, DeMed Clinic Center, Bangkok, Thailand
| | - Byong Seung Cho
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul, Korea
| | - Therdpong Tempark
- Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
7
|
Chen X, Yang N, Li B, Gao X, Wang Y, Wang Q, Liu X, Zhang Z, Zhang R. Visualization Analysis of Small Extracellular Vesicles in the Application of Bone-Related Diseases. Cells 2024; 13:904. [PMID: 38891036 PMCID: PMC11171653 DOI: 10.3390/cells13110904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Small extracellular vesicles were shown to have similar functional roles to their parent cells without the defect of potential tumorigenicity, which made them a great candidate for regenerative medicine. The last twenty years have witnessed the rapid development of research on small extracellular vesicles. In this paper, we employed a scientometric synthesis method to conduct a retrospective analysis of small extracellular vesicles in the field of bone-related diseases. The overall background analysis consisted the visualization of the countries, institutions, journals, and authors involved in research. The current status of the research direction and future trends were presented through the analysis of references and keywords, which showed that engineering strategies, mesenchymal stem cell derived exosomes, and cartilage damage were the most concerning topics, and scaffold, osteoarthritis, platelet-rich plasma, and senescence were the future trends. We also discussed the current problems and challenges in practical applications, including the in-sight mechanisms, the building of relevant animal models, and the problems in clinical trials. By using CiteSpace, VOSviewer, and Bibliometrix, the presented data avoided subjective selectivity and tendency well, which made the conclusion more reliable and comprehensive. We hope that the findings can provide new perspectives for researchers to understand the evolution of this field over time and to search for novel research directions.
Collapse
Affiliation(s)
- Xinjiani Chen
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ning Yang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
| | - Bailei Li
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xinyu Gao
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
| | - Yayu Wang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qin Wang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaojun Liu
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
- Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 318000, China
| | - Zhen Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
| | - Rongqing Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
- Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 318000, China
| |
Collapse
|
8
|
Liu D, Tang W, Tang D, Yan H, Jiao F. Ocu-miR-10a-5p promotes the chondrogenic differentiation of rabbit BMSCs by targeting BTRC-mediated Wnt/β-catenin signaling pathway. In Vitro Cell Dev Biol Anim 2024; 60:343-353. [PMID: 38504085 DOI: 10.1007/s11626-024-00888-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024]
Abstract
MicroRNAs (miRNAs) play an important role in articular cartilage damage in osteoarthritis (OA). However, the biological role of miRNAs in the chondrogenic differentiation of bone marrow mesenchymal stem cell (BMSC) remains largely unclear. Rabbit bone marrow mesenchymal stem cells (rBMSCs) were isolated, cultured, and identified. Afterwards, rBMSCs were induced to chondrogenic differentiation, examined by Alcian Blue staining. Differentially expressed miRNAs were identified in rBMSCs between induced and non-induced groups by miRNA sequencing analysis, part of which was validated via PCR assay. Cell viability and apoptosis were assessed by CCK-8 assay and Hoechst staining. Saffron O staining was utilized to assess chondrocyte hyperplasia. The expression of specific chondrogenic markers, including COL2A1, SOX9, Runx2, MMP-13, Aggrecan, and BMP-2, were measured at mRNA and protein levels. The association between beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC) and miR-10a-5p in the miRNA family from rabbit (ocu-miR-10a-5p) was determined by luciferase reporter assay. A total of 76 differentially expressed miRNAs, including 52 downregulated and 24 upregulated miRNAs, were identified in rBMSCs from the induced group. Inhibition of ocu-miR-10a-5p suppressed rBMSC viability and chondrogenic differentiation, as well as downregulated the expression of β-catenin, SOX9, COL2A1, MMP-13, and Runx2. BTRC was predicted and confirmed as a target of ocu-miR-10a-5p. Overexpression of BTRC rescued the promoting impacts of overexpressed ocu-miR-10a-5p on chondrogenic differentiation of rBMSCs and β-catenin expression. Taken together, our data suggested that ocu-miR-10a-5p facilitated rabbit BMSC survival and chondrogenic differentiation by activating Wnt/β-catenin signaling through BTRC.
Collapse
Affiliation(s)
- Donghua Liu
- Department of Spine Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Wang Tang
- Department of Spine Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Dongming Tang
- Department of Joint Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, 87 Yingbin Road, Huadu District, Guangzhou City, Guangdong Province, China
| | - Haixia Yan
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feng Jiao
- Department of Joint Surgery, Guangzhou Hospital of Integrated Traditional and Western Medicine, 87 Yingbin Road, Huadu District, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
9
|
Hussain MS, Shaikh NK, Agrawal M, Tufail M, Bisht AS, Khurana N, Kumar R. Osteomyelitis and non-coding RNAS: A new dimension in disease understanding. Pathol Res Pract 2024; 255:155186. [PMID: 38350169 DOI: 10.1016/j.prp.2024.155186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/15/2024]
Abstract
Osteomyelitis, a debilitating bone infection, presents considerable clinical challenges due to its intricate etiology and limited treatment options. Despite strides in surgical and chemotherapeutic interventions, the treatment landscape for osteomyelitis remains unsatisfactory. Recent attention has focused on the role of non-coding RNAs (ncRNAs) in the pathogenesis and progression of osteomyelitis. This review consolidates current knowledge on the involvement of distinct classes of ncRNAs, including microRNAs, long ncRNAs, and circular RNAs, in the context of osteomyelitis. Emerging evidence from various studies underscores the potential of ncRNAs in orchestrating gene expression and influencing the differentiation of osteoblasts and osteoclasts, pivotal processes in bone formation. The review initiates by elucidating the regulatory functions of ncRNAs in fundamental cellular processes such as inflammation, immune response, and bone remodeling, pivotal in osteomyelitis pathology. It delves into the intricate network of interactions between ncRNAs and their target genes, illuminating how dysregulation contributes to the establishment and persistence of osteomyelitic infections. Understanding their regulatory roles may pave the way for targeted diagnostic tools and innovative therapeutic interventions, promising a paradigm shift in the clinical approach to this challenging condition. Additionally, we delve into the promising therapeutic applications of these molecules, envisioning novel diagnostic and treatment approaches to enhance the management of this challenging bone infection.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Department of Pharmacology, School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan 302017, India
| | - Nusrat K Shaikh
- Department of Quality Assurance, Smt. N. M. Padalia Pharmacy College, Ahmedabad, 382210 Gujarat, India
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram 122103, India
| | - Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
| | - Ajay Singh Bisht
- School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand 248001, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
10
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
11
|
Zhu W, Shi J, Weng B, Zhou Z, Mao X, Pan S, Peng J, Zhang C, Mao H, Li M, Zhao J. EVs from cells at the early stages of chondrogenesis delivered by injectable SIS dECM promote cartilage regeneration. J Tissue Eng 2024; 15:20417314241268189. [PMID: 39157647 PMCID: PMC11329914 DOI: 10.1177/20417314241268189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
Articular cartilage defect therapy is still dissatisfactory in clinic. Direct cell implantation faces challenges, such as tumorigenicity, immunogenicity, and uncontrollability. Extracellular vesicles (EVs) based cell-free therapy becomes a promising alternative approach for cartilage regeneration. Even though, EVs from different cells exhibit heterogeneous characteristics and effects. The aim of the study was to discover the functions of EVs from the cells during chondrogenesis timeline on cartilage regeneration. Here, bone marrow mesenchymal stem cells (BMSCs)-EVs, juvenile chondrocytes-EVs, and adult chondrocytes-EVs were used to represent the EVs at different differentiation stages, and fibroblast-EVs as surrounding signals were also joined to compare. Fibroblasts-EVs showed the worst effect on chondrogenesis. While juvenile chondrocyte-EVs and adult chondrocyte-EVs showed comparable effect on chondrogenic differentiation as BMSCs-EVs, BMSCs-EVs showed the best effect on cell proliferation and migration. Moreover, the amount of EVs secreted from BMSCs were much more than that from chondrocytes. An injectable decellularized extracellular matrix (dECM) hydrogel from small intestinal submucosa (SIS) was fabricated as the EVs delivery platform with natural matrix microenvironment. In a rat model, BMSCs-EVs loaded SIS hydrogel was injected into the articular cartilage defects and significantly enhanced cartilage regeneration in vivo. Furthermore, protein proteomics revealed BMSCs-EVs specifically upregulated multiple metabolic and biosynthetic processes, which might be the potential mechanism. Thus, injectable SIS hydrogel loaded with BMSCs-EVs might be a promising therapeutic way for articular cartilage defect.
Collapse
Affiliation(s)
- Weilai Zhu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Jiaying Shi
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Bowen Weng
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Zhenger Zhou
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Xufeng Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Senhao Pan
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Jing Peng
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Chi Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Mei Li
- Zhejiang Key Laboratory of Precision Medicine for Atherosclerotic Diseases, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| |
Collapse
|
12
|
Iqbal Z, Xia J, Murtaza G, Shabbir M, Rehman K, Yujie L, Duan L. Targeting WNT signalling pathways as new therapeutic strategies for osteoarthritis. J Drug Target 2023; 31:1027-1049. [PMID: 37969105 DOI: 10.1080/1061186x.2023.2281861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/21/2023] [Indexed: 11/17/2023]
Abstract
Osteoarthritis (OA) is a highly prevalent chronic joint disease and the leading cause of disability. Currently, no drugs are available to control joint damage or ease the associated pain. The wingless-type (WNT) signalling pathway is vital in OA progression. Excessive activation of the WNT signalling pathway is pertinent to OA progression and severity. Therefore, agonists and antagonists of the WNT pathway are considered potential drug candidates for OA treatment. For example, SM04690, a novel small molecule inhibitor of WNT signalling, has demonstrated its potential in a recent phase III clinical trial as a disease-modifying osteoarthritis drug (DMOAD). Therefore, targeting the WNT signalling pathway may be a distinctive approach to developing particular agents helpful in treating OA. This review aims to update the most recent progress in OA drug development by targeting the WNT pathway. In this, we introduce WNT pathways and their crosstalk with other signalling pathways in OA development and highlight the role of the WNT signalling pathway as a key regulator in OA development. Several articles have reviewed the Wnt pathway from different aspects. This candid review provides an introduction to WNT pathways and their crosstalk with other signalling pathways in OA development, highlighting the role of the WNT signalling pathway as a key regulator in OA development with the latest research. Particularly, we emphasise the state-of-the-art in targeting the WNT pathway as a promising therapeutic approach for OA and challenges in their development and the nanocarrier-based delivery of WNT modulators for treating OA.
Collapse
Affiliation(s)
- Zoya Iqbal
- Department of Orthopedics, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Pakistan
| | - Maryam Shabbir
- Faculty of Pharmacy, The University of Lahore, Lahore Campus, Pakistan
| | - Khurrum Rehman
- Department of Allied health sciences, The University of Agriculture, D.I.Khan, Pakistan
| | - Liang Yujie
- Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Li Duan
- Department of Orthopedics, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Zhang H, Huang J, Alahdal M. Exosomes loaded with chondrogenic stimuli agents combined with 3D bioprinting hydrogel in the treatment of osteoarthritis and cartilage degeneration. Biomed Pharmacother 2023; 168:115715. [PMID: 37857246 DOI: 10.1016/j.biopha.2023.115715] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Osteoarthritis (OA) is a challenging joint inflammatory disease that often leads to disability. Immunoregulatory Exosomes (Exos) have shown promise in OA and cartilage degeneration treatment. Engineering Exos to deliver therapeutic agents like Kartogenin (KGN) has displayed potential for restoring cartilage regeneration. However, challenges include the uneven distribution of Exos at the injury site and the release of Exos cargo out of chondrocytes. Hydrogel-loaded uMSC-Exo has demonstrated significant therapeutic effects in wound healing and tissue regeneration. Recently, a new version of three-dimensional (3D) bioprinting of hydrogel significantly restored cartilage regeneration in OA joints. Combining immune regulatory Exos with 3D bioprinting hydrogel (3D-BPH-Exos) holds the potential for immunomodulating cartilage tissue and treatment of OA. It can reduce intracellular inflammasome formation and the release of inflammatory agents like IL-1β, TNF-α, and INF-γ, while also preventing chondrocyte apoptosis by restoring mitochondrial functions and enhancing chondrogenesis in synovial MSCs, osteoprogenitor cells, and osteoclasts. Loading Exos with chondrogenic stimuli agents in the 3D-BPH-Exos approach may offer a faster and safer strategy for cartilage repair while better inhibiting joint inflammation than high doses of anti-inflammatory drugs and cell-based therapies. This review provides a comprehensive overview of hydrogel bioprinting and exosome-based therapy in OA. It emphasizes the potential of 3D-BPH-Exos loaded with chondrogenic stimuli agents for OA treatment, serving as a basis for further research.
Collapse
Affiliation(s)
- Hui Zhang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen 518035, China; Department of Orthopedics, Shangrao People's Hospital, Shangrao, Jiangxi, China
| | - Jianghong Huang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen 518035, China.
| | - Murad Alahdal
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen 518035, China; Johns Hopkins All Children's Hospital, 600 5th St. South, St. Petersburg, FL 33701, USA.
| |
Collapse
|
14
|
Guo R, Fan J. Extracellular Vesicles Derived from Auricular Chondrocytes Facilitate Cartilage Differentiation of Adipose-Derived Mesenchymal Stem Cells. Aesthetic Plast Surg 2023; 47:2823-2832. [PMID: 36849663 DOI: 10.1007/s00266-023-03292-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/20/2023] [Indexed: 03/01/2023]
Abstract
PURPOSE Adipose-derived mesenchymal stem cell (ADSC)-based therapies have been utilized for cartilage regeneration because of their multi-lineage differentiation ability. However, commonly used cartilage inducers such as the transforming growth factor beta-3 (TGF-β3) may be prone to cartilage dedifferentiation and hypertrophy. The directional differentiation of elastic cartilage is limited nowadays. Extracellular vesicles (EVs) have been reported to influence the specific differentiation of mesenchymal stem cells (MSCs) by reflecting the composition of the parental cells. However, the role of auricular chondrogenic-derived EVs (AC-EVs) in elastic chondrogenic differentiation of ADSCs has not yet been reported. RESULTS AC-EVs isolated from the external ears of swine exhibited a positive effect on cell proliferation and migration. Furthermore, AC-EVs efficiently promoted chondrogenic differentiation of ADSCs in pellet culture, as shown by the elevated levels of COL2A1, ACAN, and SOX-9 expression. Moreover, there was a significantly higher expression of elastin and a lower expression of the fibrotic marker COL1A1 in comparison with that achieved with TGF-β3. The staining results demonstrated that AC-EVs promoted the deposition of cartilage-specific matrix, which is in good concordance with the real-time polymerase chain reaction (RT-PCR) results. CONCLUSIONS Auricular chondrogenic-derived EVs are a crucial component in elastic chondrogenic differentiation and other biological behaviors of ADSCs, which may be a useful ingredient for cartilage tissue engineering and external ear reconstruction. NO LEVEL ASSIGNED This journal requires that authors 42 assign a level of evidence to each submission to which 43 Evidence-Based Medicine rankings are applicable. This 44 excludes Review Articles, Book Reviews, and manuscripts 45 that concern Basic Science, Animal Studies, Cadaver 46 Studies, and Experimental Studies. For a full description of 47 these Evidence-Based Medicine ratings, please refer to the 48 Table oôf Contents or the online Instructions to Authors 49 www.springer.com/00266 .
Collapse
Affiliation(s)
- Rui Guo
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China
| | - Jincai Fan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 33 Badachu Road, Shijingshan District, Beijing, 100144, People's Republic of China.
| |
Collapse
|
15
|
Li Z, Bi R, Zhu S. The Dual Role of Small Extracellular Vesicles in Joint Osteoarthritis: Their Global and Non-Coding Regulatory RNA Molecule-Based Pathogenic and Therapeutic Effects. Biomolecules 2023; 13:1606. [PMID: 38002288 PMCID: PMC10669328 DOI: 10.3390/biom13111606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
OA is the most common joint disease that affects approximately 7% of the global population. Current treatment methods mainly relieve its symptoms with limited repairing effect on joint destructions, which ultimately contributes to the high morbidity rate of OA. Stem cell treatment is a potential regenerative medical therapy for joint repair in OA, but the uncertainty in differentiation direction and immunogenicity limits its clinical usage. Small extracellular vesicles (sEVs), the by-products secreted by stem cells, show similar efficacy levels but have safer regenerative repair effect without potential adverse outcomes, and have recently drawn attention from the broader research community. A series of research works and reviews have been performed in the last decade, providing references for the application of various exogenous therapeutic sEVs for treating OA. However, the clinical potential of target intervention involving endogenous pathogenic sEVs in the treatment of OA is still under-explored and under-discussed. In this review, and for the first time, we emphasize the dual role of sEVs in OA and explain the effects of sEVs on various joint tissues from both the pathogenic and therapeutic aspects. Our aim is to provide a reference for future research in the field.
Collapse
Affiliation(s)
- Zhi Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Ruiye Bi
- Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Songsong Zhu
- Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
16
|
Deng S, Cao H, Cui X, Fan Y, Wang Q, Zhang X. Optimization of exosome-based cell-free strategies to enhance endogenous cell functions in tissue regeneration. Acta Biomater 2023; 171:68-84. [PMID: 37730080 DOI: 10.1016/j.actbio.2023.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Exosomes, nanoscale extracellular vesicles, play a crucial role in intercellular communication, owing to their biologically active cargoes such as RNAs and proteins. In recent years, they have emerged as a promising tool in the field of tissue regeneration, with the potential to initiate a new trend in cell-free therapy. However, it's worth noting that not all types of exosomes derived from cells are appropriate for tissue repair. Thus, selecting suitable cell sources is critical to ensure their efficacy in specific tissue regeneration processes. Current therapeutic applications of exosomes also encounter several limitations, including low-specific content for targeted diseases, non-tissue-specific targeting, and short retention time due to rapid clearance in vivo. Consequently, this review paper focuses on exosomes from diverse cell sources with functions specific to tissue regeneration. It also highlights the latest engineering strategies developed to overcome the functional limitations of natural exosomes. These strategies encompass the loading of specific therapeutic contents into exosomes, the endowment of tissue-specific targeting capability on the exosome surface, and the incorporation of biomaterials to extend the in vivo retention time of exosomes in a controlled-release manner. Collectively, these innovative approaches aim to synergistically enhance the therapeutic effects of natural exosomes, optimizing exosome-based cell-free strategies to boost endogenous cell functions in tissue regeneration. STATEMENT OF SIGNIFICANCE: Exosome-based cell-free therapy has recently emerged as a promising tool for tissue regeneration. This review highlights the characteristics and functions of exosomes from different sources that can facilitate tissue repair and their contributions to the regeneration process. To address the functional limitations of natural exosomes in therapeutic applications, this review provides an in-depth understanding of the latest engineering strategies. These strategies include optimizing exosomal contents, endowing tissue-specific targeting capability on the exosome surface, and incorporating biomaterials to extend the in vivo retention time of exosomes in a controlled-release manner. This review aims to explore and discuss innovative approaches that can synergistically improve endogenous cell functions in advanced exosome-based cell-free therapies for a broad range of tissue regeneration.
Collapse
Affiliation(s)
- Siyan Deng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hongfu Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaolin Cui
- School of medicine, the Chinese University of Hong Kong, Shenzhen, China; Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
17
|
Wang Y, Liu Z, Pan C, Zheng Y, Chen Y, Lian X, Jiang Y, Chen C, Xue K, Zhang Y, Xu P, Liu K. Ultrasound-Driven Healing: Unleashing the Potential of Chondrocyte-Derived Extracellular Vesicles for Chondrogenesis in Adipose-Derived Stem Cells. Biomedicines 2023; 11:2836. [PMID: 37893208 PMCID: PMC10604747 DOI: 10.3390/biomedicines11102836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Repairing cartilage defects represents a significant clinical challenge. While adipose-derived stem cell (ADSC)-based strategies hold promise for cartilage regeneration, their inherent chondrogenic potential is limited. Extracellular vesicles (EVs) derived from chondrocytes (CC-EVs) have shown potential in enhancing chondrogenesis, but their role in promoting chondrogenic differentiation of ADSCs remains poorly understood. Moreover, the clinical application of EVs faces limitations due to insufficient quantities for in vivo use, necessitating the development of effective methods for extracting significant amounts of CC-EVs. Our previous study demonstrated that low-intensity ultrasound (LIUS) stimulation enhances EV secretion from mesenchymal stem cells. Here, we identified a specific LIUS parameter for chondrocytes that increased EV secretion by 16-fold. CC-EVs were found to enhance cell activity, proliferation, migration, and 21-day chondrogenic differentiation of ADSCs in vitro, while EVs secreted by chondrocytes following LIUS stimulation (US-CC-EVs) exhibited superior efficacy. miRNA-seq revealed that US-CC-EVs were enriched in cartilage-regeneration-related miRNAs, contributing to chondrogenesis in various biological processes. In conclusion, we found that CC-EVs can enhance the chondrogenesis of ADSCs in vitro. In addition, our study introduces ultrasound-driven healing as an innovative method to enhance the quantity and quality of CC-EVs, meeting clinical demand and addressing the limited chondrogenic potential of ADSCs. The ultrasound-driven healing unleashes the potential of CC-EVs for chondrogenesis possibly through the enrichment of cartilage-regeneration-associated miRNAs in EVs, suggesting their potential role in cartilage reconstruction. These findings hold promise for advancing cartilage regeneration strategies and may pave the way for novel therapeutic interventions in regenerative medicine.
Collapse
Affiliation(s)
- Yikai Wang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Zibo Liu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Chuqiao Pan
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yi Zheng
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yahong Chen
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Xiang Lian
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yu Jiang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Chuhsin Chen
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Ke Xue
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27101, USA;
| | - Peng Xu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Kai Liu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| |
Collapse
|
18
|
Ryu Y, Hwang JS, Bo Noh K, Park SH, Seo JH, Shin YJ. Adipose Mesenchymal Stem Cell-Derived Exosomes Promote the Regeneration of Corneal Endothelium Through Ameliorating Senescence. Invest Ophthalmol Vis Sci 2023; 64:29. [PMID: 37850944 PMCID: PMC10593138 DOI: 10.1167/iovs.64.13.29] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Purpose Human corneal endothelial cells (hCECs) have been considered unable to regenerate in vivo, resulting in corneal decompensation after significant loss of hCECs. adipose-derived mesenchymal stem cell (ASC)-derived exosomes can regenerate tissues and organs. In this study, we investigated whether ASC-derived exosomes could protect and regenerate CECs. Methods We performed cell viability and cell-cycle analyses to evaluate the effect of ASC-derived exosomes on the regeneration capacity of cultured hCECs. Transforming growth factor-β (TGF-β) and hydrogen peroxide (H2O2) were used to induce biological stress in CECs. The effect of ASC-derived exosomes on CECs was investigated in vivo. ASC-derived exosomes were introduced into rat CECs using electroporation, and rat corneas were injured using cryoinjury. Next-generation sequencing analysis was performed to compare the differentially expressed microRNAs (miRNAs) between ASC-derived and hCEC-derived exosomes. Results ASC-derived exosomes induced CEC proliferation and suppressed TGF-β- or H2O2-induced oxidative stress and senescence. ASC-derived exosomes protect hCECs against TGF-β- or H2O2-induced endothelial-mesenchymal transition and mitophagy. In an in vivo study, ASC-derived exosomes promoted wound healing of rat CECs and protected the corneal endothelium against cryoinjury-induced corneal endothelium damage. Next-generation sequencing analysis revealed differentially expressed miRNAs for ASC-derived and hCEC-derived exosomes. They are involved in lysine degradation, adherens junction, the TGF-β signaling pathway, the p53 signaling pathway, the Hippo signaling pathway, the forkhead box O (FoxO) signaling pathway, regulation of actin cytoskeleton, and RNA degradation based on Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Conclusions ASC-derived exosomes promoted wound healing and regeneration of endothelial cells by inducing a shift in the cell cycle and suppressing senescence and autophagy.
Collapse
Affiliation(s)
- Yunkyoung Ryu
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Kyung Bo Noh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Se Hie Park
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
19
|
Yin P, Jiang Y, Fang X, Wang D, Li Y, Chen M, Deng H, Tang P, Zhang L. Cell-Based Therapies for Degenerative Musculoskeletal Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207050. [PMID: 37199688 PMCID: PMC10375105 DOI: 10.1002/advs.202207050] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/29/2023] [Indexed: 05/19/2023]
Abstract
Degenerative musculoskeletal diseases (DMDs), including osteoporosis, osteoarthritis, degenerative disc disease, and sarcopenia, present major challenges in the aging population. Patients with DMDs present with pain, functional decline, and reduced exercise tolerance, which result in long-term or permanent deficits in their ability to perform daily activities. Current strategies for dealing with this cluster of diseases focus on relieving pain, but they have a limited capacity to repair function or regenerate tissue. Cell-based therapies have attracted considerable attention in recent years owing to their unique mechanisms of action and remarkable effects on regeneration. In this review, current experimental attempts to use cell-based therapies for DMDs are highlighted, and the modes of action of different cell types and their derivatives, such as exosomes, are generalized. In addition, the latest findings from state-of-the-art clinical trials are reviewed, approaches to improve the efficiency of cell-based therapies are summarized, and unresolved questions and potential future research directions for the translation of cell-based therapies are identified.
Collapse
Affiliation(s)
- Pengbin Yin
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Yuheng Jiang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
- Department of OrthopedicsGeneral Hospital of Southern Theater Command of PLANo. 111, Liuhua AvenueGuangzhou510010China
| | - Xuan Fang
- Department of Anatomy, Histology and EmbryologySchool of Basic Medical SciencesPeking University Health Science CenterBeijing100191China
| | - Daofeng Wang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Yi Li
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Ming Chen
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Hao Deng
- Department of OrthopedicsThird Affiliated Hospital of Jinzhou Medical UniversityJinzhouLiaoning Province121000China
| | - Peifu Tang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| | - Licheng Zhang
- Department of Orthopedicsthe Fourth Medical CenterChinese PLA General HospitalBeijing100853China
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationBeijing100853China
| |
Collapse
|
20
|
Cong B, Sun T, Zhao Y, Chen M. Current and Novel Therapeutics for Articular Cartilage Repair and Regeneration. Ther Clin Risk Manag 2023; 19:485-502. [PMID: 37360195 PMCID: PMC10290456 DOI: 10.2147/tcrm.s410277] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
Articular cartilage repair is a sophisticated process that has is being recently investigated. There are several different approaches that are currently reported to promote cartilage repair, like cell-based therapies, biologics, and physical therapy. Cell-based therapies involve the using stem cells or chondrocytes, which make up cartilage, to promote the growth of new cartilage. Biologics, like growth factors, are also being applied to enhance cartilage repair. Physical therapy, like exercise and weight-bearing activities, can also be used to promote cartilage repair by inducing new cartilage growth and improving joint function. Additionally, surgical options like osteochondral autograft, autologous chondrocyte implantation, microfracture, and others are also reported for cartilage regeneration. In the current literature review, we aim to provide an up-to-date discussion about these approaches and discuss the current research status.
Collapse
Affiliation(s)
- Bo Cong
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Tao Sun
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Yuchi Zhao
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Mingqi Chen
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
| |
Collapse
|
21
|
Wawrzkiewicz-Jałowiecka A, Lalik A, Lukasiak A, Richter-Laskowska M, Trybek P, Ejfler M, Opałka M, Wardejn S, Delfino DV. Potassium Channels, Glucose Metabolism and Glycosylation in Cancer Cells. Int J Mol Sci 2023; 24:ijms24097942. [PMID: 37175655 PMCID: PMC10178682 DOI: 10.3390/ijms24097942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Potassium channels emerge as one of the crucial groups of proteins that shape the biology of cancer cells. Their involvement in processes like cell growth, migration, or electric signaling, seems obvious. However, the relationship between the function of K+ channels, glucose metabolism, and cancer glycome appears much more intriguing. Among the typical hallmarks of cancer, one can mention the switch to aerobic glycolysis as the most favorable mechanism for glucose metabolism and glycome alterations. This review outlines the interconnections between the expression and activity of potassium channels, carbohydrate metabolism, and altered glycosylation in cancer cells, which have not been broadly discussed in the literature hitherto. Moreover, we propose the potential mediators for the described relations (e.g., enzymes, microRNAs) and the novel promising directions (e.g., glycans-orinented drugs) for further research.
Collapse
Affiliation(s)
- Agata Wawrzkiewicz-Jałowiecka
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Anna Lalik
- Department of Systems Biology and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Agnieszka Lukasiak
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Monika Richter-Laskowska
- The Centre for Biomedical Engineering, Łukasiewicz Research Network-Krakow Institute of Technology, 30-418 Krakow, Poland
| | - Paulina Trybek
- Institute of Physics, University of Silesia in Katowice, 41-500 Chorzów, Poland
| | - Maciej Ejfler
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Maciej Opałka
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Sonia Wardejn
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Domenico V Delfino
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
| |
Collapse
|
22
|
Mishra A, Kumar R, Mishra SN, Vijayaraghavalu S, Tiwari NK, Shukla GC, Gurusamy N, Kumar M. Differential Expression of Non-Coding RNAs in Stem Cell Development and Therapeutics of Bone Disorders. Cells 2023; 12:cells12081159. [PMID: 37190068 PMCID: PMC10137108 DOI: 10.3390/cells12081159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Stem cells' self-renewal and multi-lineage differentiation are regulated by a complex network consisting of signaling factors, chromatin regulators, transcription factors, and non-coding RNAs (ncRNAs). Diverse role of ncRNAs in stem cell development and maintenance of bone homeostasis have been discovered recently. The ncRNAs, such as long non-coding RNAs, micro RNAs, circular RNAs, small interfering RNA, Piwi-interacting RNAs, etc., are not translated into proteins but act as essential epigenetic regulators in stem cells' self-renewal and differentiation. Different signaling pathways are monitored efficiently by the differential expression of ncRNAs, which function as regulatory elements in determining the fate of stem cells. In addition, several species of ncRNAs could serve as potential molecular biomarkers in early diagnosis of bone diseases, including osteoporosis, osteoarthritis, and bone cancers, ultimately leading to the development of new therapeutic strategies. This review aims to explore the specific roles of ncRNAs and their effective molecular mechanisms in the growth and development of stem cells, and in the regulation of osteoblast and osteoclast activities. Furthermore, we focus on and explore the association of altered ncRNA expression with stem cells and bone turnover.
Collapse
Affiliation(s)
- Anurag Mishra
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Rishabh Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Satya Narayan Mishra
- Maa Gayatri College of Pharmacy, Dr. APJ Abdul Kalam Technical University, Prayagraj 211009, India
| | | | - Neeraj Kumar Tiwari
- Department of IT-Satellite Centre, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India
| | - Girish C Shukla
- Department of Biological, Geological, and Environmental Sciences, 2121 Euclid Ave., Cleveland, OH 44115, USA
- Center for Gene Regulation in Health and Disease, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Munish Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| |
Collapse
|
23
|
Li S, Wu C, Lin S, Wen Z, Luo W, Li C, Wang X, Li X, Gao L, Ding Y. HUCMSC-derived Exosomes Suppress the Titanium Particles-induced Osteolysis in Mice through Inhibiting CCL2 and CCL3. Orthop Surg 2023; 15:888-898. [PMID: 36720704 PMCID: PMC9977603 DOI: 10.1111/os.13608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE Wear particles induce inflammation and the further osteolysis around the prosthesis, has been proven to be the main cause of aseptic hip joint loosening. In this research, we aimed to clarify whether human umbilical cord mesenchymal stem cells (HUCMSCs) could inhibit the titanium particles-induced osteolysis and shed light upon its mechanism. METHODS The expression of chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-C motif) ligand 3 (CCL3) and chemokine (C-C motif) ligand 5 (CCL5) were examinjed in clinical specimens of aseptic hip prosthesis loosening patients. Local injection of lentivirus that knocked down CCL2 or CCL3 in a cranial osteolysis mice model were used to exam the effect of CCL2 and CCL3 on titanium particles-induced osteolysis in vivo. Transwell assay was used to examine the effect of CCL2 and CCL3 on titanium particles-induced activation of macrophage in vitro. Furthermore, the therapeutic effect of HUCMSCs, and exosomes from HUCMSCs were also examed in vivo and vitro. Immunohistochemical and real-time PCR were used to examine the expression of relative pathways. Analysis of variance (ANOVA) and Student-Newman-Keuls post hoc t test were used to analyze the results and determine the statistical significance of the differences. RESULTS Results showed that titanium particles caused the osteolysis at the mice cranial in vivo and a large number of macrophages that migrated, while local injection of HUCMSCs and exosomes did inhibit the cranial osteolysis and migration. An exosome inhibitor GW4869 significantly increased the osteolysis area in the mice cranium osteolysis model, and increased the number of migrated macrophages. Immunohistochemical results suggested that the expression of CCL2, CCL3 and CD68 in the cranial in Titanium particles mice increased significantly, but was significantly reduced by HUCMSCs or exosomes. HUCMSC and exosomes down-regulate the expression of CCL3 in vitro and in vivo. CONCLUSION HUCMSCs and HUCMSC-derived exosomes could suppress the titanium particles-induced osteolysis in mice through inhibiting chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 3.
Collapse
Affiliation(s)
- Shixun Li
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Chuangran Wu
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Sipeng Lin
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Zhenkang Wen
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Wenqiang Luo
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Changchuan Li
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Xiaoyan Wang
- Guangzhou Saliai Stem Cell Science and Technology Co., LTDGuangzhouChina
| | - Xuejia Li
- Guangzhou Saliai Stem Cell Science and Technology Co., LTDGuangzhouChina
| | - Liangbin Gao
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yue Ding
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
24
|
Liu F, Sun T, An Y, Ming L, Li Y, Zhou Z, Shang F. The potential therapeutic role of extracellular vesicles in critical-size bone defects: Spring of cell-free regenerative medicine is coming. Front Bioeng Biotechnol 2023; 11:1050916. [PMID: 36733961 PMCID: PMC9887316 DOI: 10.3389/fbioe.2023.1050916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
In recent years, the incidence of critical-size bone defects has significantly increased. Critical-size bone defects seriously affect patients' motor functions and quality of life and increase the need for additional clinical treatments. Bone tissue engineering (BTE) has made great progress in repairing critical-size bone defects. As one of the main components of bone tissue engineering, stem cell-based therapy is considered a potential effective strategy to regenerate bone tissues. However, there are some disadvantages including phenotypic changes, immune rejection, potential tumorigenicity, low homing efficiency and cell survival rate that restrict its wider clinical applications. Evidence has shown that the positive biological effects of stem cells on tissue repair are largely mediated through paracrine action by nanostructured extracellular vesicles (EVs), which may overcome the limitations of traditional stem cell-based treatments. In addition to stem cell-derived extracellular vesicles, the potential therapeutic roles of nonstem cell-derived extracellular vesicles in critical-size bone defect repair have also attracted attention from scholars in recent years. Currently, the development of extracellular vesicles-mediated cell-free regenerative medicine is still in the preliminary stage, and the specific mechanisms remain elusive. Herein, the authors first review the research progress and possible mechanisms of extracellular vesicles combined with bone tissue engineering scaffolds to promote bone regeneration via bioactive molecules. Engineering modified extracellular vesicles is an emerging component of bone tissue engineering and its main progression and clinical applications will be discussed. Finally, future perspectives and challenges of developing extracellular vesicle-based regenerative medicine will be given. This review may provide a theoretical basis for the future development of extracellular vesicle-based biomedicine and provide clinical references for promoting the repair of critical-size bone defects.
Collapse
Affiliation(s)
- Fen Liu
- Department of Periodontology, Shenzhen Stomatological Hospital (Pingshan), Southern Medical University, Shenzhen, Guangdong, China
| | - Tianyu Sun
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying An
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture and Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Leiguo Ming
- Department of Research and Development, Shaanxi Zhonghong Institute of Regenerative Medicine, Xi’an, Shaanxi, China
| | - Yinghui Li
- Department of Orthodontics, Stomatological Hospital, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, Tibet, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| | - Fengqing Shang
- Department of Stomatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China,*Correspondence: Fengqing Shang, ; Zhifei Zhou,
| |
Collapse
|
25
|
Bai Z, Hu K, Shou Z, Yu J, Meng H, Zhou H, Chen L, Yu T, Lu R, Li N, Chen C. Layer-by-layer assembly of procyanidin and collagen promotes mesenchymal stem cell proliferation and osteogenic differentiation in vitro and in vivo. Regen Biomater 2022; 10:rbac107. [PMID: 36683760 PMCID: PMC9847536 DOI: 10.1093/rb/rbac107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/13/2022] [Accepted: 12/09/2022] [Indexed: 12/27/2022] Open
Abstract
Collagen, commonly used in tissue engineering, is widespread in various tissues. During bone tissue regeneration, collagen can stimulate the cellular response and determine the fate of cells. In this work, we integrated collagen type II with procyanidin (PC) onto an implant coating by applying a layer-by-layer technique to demonstrate that collagen and PC can participate in the construction of new biomaterials and serve as multifunctional components. The effects of PC/collagen multilayers on the viability of cocultured bone marrow mesenchymal stem cells (BMSCs) were analyzed by cell counting kit-8 analysis and phalloidin staining. The reactive oxygen species level of BMSCs was revealed through immunofluorescent staining and flow cytometry. Osteogenesis-related genes were detected, and in vivo experiment was performed to reveal the effect of newly designed material on the osteogenic differentiation of BMSCs. Our data demonstrated that in BMSCs PC/collagen multilayers accelerated the proliferation and osteogenic differentiation through Wnt/β-catenin signaling pathway and enhanced bone generation around the implant in the bone defect model of rabbit femurs. In summary, combination of collagen and PC provided a new sight for the research and development of implant materials or coatings in the future.
Collapse
Affiliation(s)
- Zhibiao Bai
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, P.R. China.,Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Kai Hu
- Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Zeyu Shou
- Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Jiahuan Yu
- Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Hongming Meng
- Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Han Zhou
- Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Liangyan Chen
- Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Tiantian Yu
- Wenzhou Key Laboratory of Perioperative Medicine, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, P.R. China.,Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, P.R. China
| | - Ruofei Lu
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, P.R. China
| | - Na Li
- Wenzhou Key Laboratory of Perioperative Medicine, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, P.R. China
| | - Chun Chen
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, P.R. China.,Wenzhou Medical University, Wenzhou 325000, P.R. China
| |
Collapse
|
26
|
Shi Y, Shao J, Zhang Z, Zhang J, Lu H. Effect of condylar chondrocyte exosomes on condylar cartilage osteogenesis in rats under tensile stress. Front Bioeng Biotechnol 2022; 10:1061855. [PMID: 36561044 PMCID: PMC9766957 DOI: 10.3389/fbioe.2022.1061855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Functional orthoses are commonly used to treat skeletal Class II malocclusion, but the specific mechanism through which they do this has been a challenging topic in orthodontics. In the present study, we aimed to explore the effect of tensile stress on the osteogenic differentiation of condylar chondrocytes from an exosomal perspective. Methods: We cultured rat condylar chondrocytes under resting and tensile stress conditions and subsequently extracted cellular exosomes from them. We then screened miRNAs that were differentially expressed between the two exosome extracts by high-throughput sequencing and performed bioinformatics analysis and osteogenesis-related target gene prediction using the TargetScan and miRanda softwares. Exosomes cultured under resting and tensile stress conditions were co-cultured with condylar chondrocytes for 24 h to form the Control-Exo and Force-Exo exosome groups, respectively. Quantitative real time PCR(RT-qPCR) and western blotting were then used to determine the mRNA and protein expression levels of Runx2 and Sox9 in condylar chondrocytes. Results: The mRNA and protein expression levels of Runx2 and Sox9 in the Force-Exo group were significantly higher than those in the Control-Exo group (p < 0.05). The differential miRNA expression results were consistent with our sequencing results. Bioinformatics analysis and target gene prediction results showed that the main biological processes and molecular functions involved in differential miRNA expression in exosomes under tensile stress were biological processes and protein binding, respectively. Kyoto Gene and Genome Data Bank (KEGG) pathway enrichment analysis showed significant enrichment of differentially expressed miRNAs in the mTOR signaling pathway. The differentially expressed miRNAs were found to target osteogenesis-related genes. Conclusion: These results suggest that stimulation of rat condylar chondrocytes with tensile stress can alter the expression levels of certain miRNAs in their exosomes and promote their osteogenic differentiation. Exosomes under tensile stress culture conditions thus have potential applications in the treatment of Osteoarthritis (OA).
Collapse
Affiliation(s)
- Yuan Shi
- Department of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Shao
- Department of Stomatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Zanzan Zhang
- Department of Stomatology, Ningbo No. 2 Hospital, Ningbo, China
| | - Jianan Zhang
- Department of Dentistry, Center of Orthodontics, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Haiping Lu
- Department of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China,*Correspondence: Haiping Lu,
| |
Collapse
|
27
|
Wang W, Liang X, Zheng K, Ge G, Chen X, Xu Y, Bai J, Pan G, Geng D. Horizon of exosome-mediated bone tissue regeneration: The all-rounder role in biomaterial engineering. Mater Today Bio 2022; 16:100355. [PMID: 35875196 PMCID: PMC9304878 DOI: 10.1016/j.mtbio.2022.100355] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/02/2022] Open
Abstract
Bone injury repair has always been a tricky problem in clinic, the recent emergence of bone tissue engineering provides a new direction for the repair of bone injury. However, some bone tissue processes fail to achieve satisfactory results mainly due to insufficient vascularization or cellular immune rejection. Exosomes with the ability of vesicle-mediated intercellular signal transmission have gained worldwide attention and can achieve cell-free therapy. Exosomes are small vesicles that are secreted by cells, which contain genetic material, lipids, proteins and other substances. It has been found to play the function of material exchange between cells. It is widely used in bone tissue engineering to achieve cell-free therapy because it not only does not produce some immune rejection like cells, but also can play a cell-like function. Exosomes from different sources can bind to scaffolds in various ways and affect osteoblast, angioblast, and macrophage polarization in vivo to promote bone regeneration. This article reviews the recent research progress of exosome-loaded tissue engineering, focusing on the mechanism of exosomes from different sources and the application of exosome-loaded scaffolds in promoting bone regeneration. Finally, the existing deficiencies and challenges, future development directions and prospects are summarized.
Collapse
Affiliation(s)
- Wentao Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Xiaolong Liang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Kai Zheng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| |
Collapse
|
28
|
Huang W, Wu X, Xiang S, Qiao M, Li H, Zhu Y, Zhu Z, Zhao Z. Regulatory of miRNAs in tri-lineage differentiation of C3H10T1/2. Stem Cell Res Ther 2022; 13:521. [PMID: 36414991 PMCID: PMC9682817 DOI: 10.1186/s13287-022-03205-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/28/2022] [Indexed: 11/24/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding single-stranded RNA molecules encoded by endogenous genes, which play a vital role in cell generation, metabolism, apoptosis and stem cell differentiation. C3H10T1/2, a mesenchymal cell extracted from mouse embryos, is capable of osteogenic differentiation, adipogenic differentiation and chondrogenic differentiation. Extensive studies have shown that not only miRNAs can directly trigger targeted genes to regulate the tri-lineage differentiation of C3H10T1/2, but it also can indirectly regulate the differentiation by triggering different signaling pathways or various downstream molecules. This paper aims to clarify the regulatory roles of different miRNAs on C3H10T1/2 differentiation, and discussing their balance effect among osteogenic differentiation, adipogenic differentiation and chondrogenic differentiation of C3H10T1/2. We also review the biogenesis of miRNAs, Wnt signaling pathways, MAPK signaling pathways and BMP signaling pathways and provide some specific examples of how these signaling pathways act on C3H10T1/2 tri-lineage differentiation. On this basis, we hope that a deeper understanding of the differentiation and regulation mechanism of miRNAs in C3H10T1/2 can provide a promising therapeutic method for the clinical treatment of bone defects, osteoporosis, osteoarthritis and other diseases.
Collapse
Affiliation(s)
- Wei Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xiaoyue Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Shuaixi Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Mingxin Qiao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Hanfei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yujie Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
29
|
Esmaeili A, Hosseini S, Kamali A, Hosseinzadeh M, Shekari F, Baghaban Eslaminejad M. Co-aggregation of MSC/chondrocyte in a dynamic 3D culture elevates the therapeutic effect of secreted extracellular vesicles on osteoarthritis in a rat model. Sci Rep 2022; 12:19827. [PMID: 36400827 PMCID: PMC9674636 DOI: 10.1038/s41598-022-22592-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular vesicles (EVs) have therapeutic effects on osteoarthritis (OA). Some recent strategies could elevate EV's therapeutic properties including cell aggregation, co-culture, and 3D culture. It seems that a combination of these strategies could augment EV production and therapeutic potential. The current study aims to evaluate the quantity of EV yield and the therapeutic effect of EVs harvested from rabbit mesenchymal stem cells (MSCs) aggregates, chondrocyte aggregates, and their co-aggregates in a dynamic 3D culture in a rat osteoarthritis model. MSC and chondrocytes were aggregated and co-aggregated by spinner flasks, and their conditioned medium was collected. EVs were isolated by size exclusion chromatography and characterized in terms of size, morphology and surface markers. The chondrogenic potential of the MSC-ag, Cho-ag and Co-ag EVs on MSC micromass differentiation in chondrogenic media were assessed by qRT-PCR, histological and immunohistochemical analysis. 50 μg of MSC-ag-EVs, Cho-ag-EVs and Co-ag-EVs was injected intra-articularly per knee of OA models established by monoiodoacetate in rats. After 8 weeks follow up, the knee joints were harvested and analyzed by radiographic, histological and immunohistochemical features. MSC/chondrocyte co-aggregation in comparison to MSC or chondrocyte aggregation could increase EV yield during dynamic 3D culture by spinner flasks. Although MSC-ag-, Cho-ag- and Co-ag-derived EVs could induce chondrogenesis similar to transforming growth factor-beta during in vitro study, Co-ag-EV could more effectively prevent OA progression than MSC-ag- and Cho-ag-EVs. Our study demonstrated that EVs harvested from the co-aggregation of MSCs and chondrocytes could be considered as a new therapeutic potential for OA treatment.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Kamali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Hosseinzadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
30
|
Kurenkova AD, Romanova IA, Kibirskiy PD, Timashev P, Medvedeva EV. Strategies to Convert Cells into Hyaline Cartilage: Magic Spells for Adult Stem Cells. Int J Mol Sci 2022; 23:11169. [PMID: 36232468 PMCID: PMC9570095 DOI: 10.3390/ijms231911169] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/30/2022] Open
Abstract
Damaged hyaline cartilage gradually decreases joint function and growing pain significantly reduces the quality of a patient's life. The clinically approved procedure of autologous chondrocyte implantation (ACI) for treating knee cartilage lesions has several limits, including the absence of healthy articular cartilage tissues for cell isolation and difficulties related to the chondrocyte expansion in vitro. Today, various ACI modifications are being developed using autologous chondrocytes from alternative sources, such as the auricles, nose and ribs. Adult stem cells from different tissues are also of great interest due to their less traumatic material extraction and their innate abilities of active proliferation and chondrogenic differentiation. According to the different adult stem cell types and their origin, various strategies have been proposed for stem cell expansion and initiation of their chondrogenic differentiation. The current review presents the diversity in developing applied techniques based on autologous adult stem cell differentiation to hyaline cartilage tissue and targeted to articular cartilage damage therapy.
Collapse
Affiliation(s)
- Anastasiia D. Kurenkova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
| | - Irina A. Romanova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Pavel D. Kibirskiy
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Ekaterina V. Medvedeva
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
| |
Collapse
|
31
|
Engineered extracellular vesicles: Regulating the crosstalk between the skeleton and immune system. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
32
|
Wu R, Li H, Sun C, Liu J, Chen D, Yu H, Huang Z, Lin S, Chen Y, Zheng Q. Exosome-based strategy for degenerative disease in orthopedics: Recent progress and perspectives. J Orthop Translat 2022; 36:8-17. [PMID: 35891923 PMCID: PMC9283806 DOI: 10.1016/j.jot.2022.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/07/2022] [Accepted: 05/19/2022] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Degenerative diseases in orthopaedics have become a significant global public health issue with the aging of the population worldwide. The traditional medical interventions, including physical therapy, pharmacological therapy and even surgery, hardly work to modify degenerative progression. Stem cell-based therapy is widely accepted to treat degenerative orthopaedic disease effectively but possesses several limitations, such as the need for strict monitoring of production and storage and the potential risks of tumorigenicity and immune rejection in clinical translation. Furthermore, the ethical issues surrounding the acquisition of embryonic stem cells are also broadly concerned. Exosome-based therapy has rapidly grown in popularity in recent years and is regarded as an ideal alternative to stem cell-based therapy, offering a promise to achieve 'cell-free' tissue regeneration. METHODS Traditionally, the native exosomes extracted from stem cells are directly injected into the injured site to promote tissue regeneration. Recently, several modified exosome-based strategies were developed to overcome the limitations of native exosomes, which include mainly exogenous molecule loading and exosome delivery through scaffolds. In this paper, a systematic review of the exosome-based strategy for degenerative disease in orthopaedics is presented. RESULTS Treatment strategies based on the native exosomes are effective but with several disadvantages such as rapid diffusion and insufficient and fluctuating functional contents. The modified exosome-based strategies can better match the requirements of the regeneration in some complex healing processes. CONCLUSION Exosome-based strategies hold promise to manage degenerative disease in orthopaedics prior to patients reaching the advanced stage of disease in the future. The timely summary and highlights offered herein could provide a research perspective to promote the development of exosome-based therapy, facilitating the clinical translation of exosomes in orthopaedics. TRANSLATIONAL POTENTIAL OF THIS ARTICLE Exosome-based therapy is superior in anti-senescence and anti-inflammatory effects and possesses lower risks of tumorigenicity and immune rejection relative to stem cell-based therapy. Exosome-based therapy is regarded as an ideal alternative to stem cell-based therapy, offering a promise to achieve 'cell-free' tissue regeneration.
Collapse
Affiliation(s)
- Rongjie Wu
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Shantou University Medical College, Shantou, China
| | - Haotao Li
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Shantou University Medical College, Shantou, China
| | - Chuanwei Sun
- Department of Burn and Wound Repair Surgery and Research Department of Medical Science, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Jialin Liu
- Rehabilitation Center, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, PR China
| | - Duanyong Chen
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Haiyang Yu
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Zena Huang
- Department of General Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
| | - Yuanfeng Chen
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Research Department of Medical Science, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Southern Medical University, Guangzhou, PR China
| |
Collapse
|
33
|
Lv B, Cheng Z, Yu Y, Chen Y, Gan W, Li S, Zhao K, Yang C, Zhang Y. Therapeutic perspectives of exosomes in glucocorticoid-induced osteoarthrosis. Front Surg 2022; 9:836367. [PMID: 36034358 PMCID: PMC9405187 DOI: 10.3389/fsurg.2022.836367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022] Open
Abstract
Exosomes are widely involved in a variety of physiological and pathological processes. These important roles are also hidden in the physiological processes related to bone. Chondrocytes, osteoblasts, synovial fibroblasts, and bone marrow mesenchymal stem cells produce and secrete exosomes, thereby affecting the biology process of target cells. Furthermore, in the primary pathogenesis of osteoarthrosis induced by steroid hormones, mainly involve glucocorticoid (GC), the exosomes have also widely participated. Therefore, exosomes may also play an important role in glucocorticoid-induced osteoarthrosis and serve as a promising treatment for early intervention of osteoarthrosis in addition to playing a regulatory role in malignant tumors. This review summarizes the previous results on this direction, systematically combs the role and therapeutic potential of exosomes in GC-induced osteoarthrosis, discusses the potential role of exosomes in the treatment and prevention of GC-induced osteoarthrosis, and reveals the current challenges we confronted.
Collapse
Affiliation(s)
- Bin Lv
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| | | | | | | | | | | | - Kangcheng Zhao
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| | - Cao Yang
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| | - Yukun Zhang
- Correspondence: Yukun ZhangCao Yang Kangcheng Zhao Bin Lv
| |
Collapse
|
34
|
Luo X, Xu Y, Zhong Z, Xiang P, Wu X, Chong A. miR-8485 alleviates the injury of cardiomyocytes through TP53INP1. J Biochem Mol Toxicol 2022; 36:e23159. [PMID: 35876212 DOI: 10.1002/jbt.23159] [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: 09/06/2021] [Revised: 04/25/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022]
Abstract
MicroRNAs (miRNAs) feature prominently in regulating the progression of chronic heart failure (CHF). This study was performed to investigate the role of miR-8485 in the injury of cardiomyocytes and CHF. It was found that miR-8485 level was markedly reduced in the plasma of CHF patients, compared with the healthy controls. H2 O2 treatment increased tumor necrosis factor-α, interleukin (IL)-6, and IL-1β levels, inhibited the viability of human adult ventricular cardiomyocyte cell line AC16, and increased the apoptosis, while miR-8485 overexpression reversed these effects. Tumor protein p53 inducible nuclear protein 1 (TP53INP1) was identified as a downstream target of miR-8485, and TP53INP1 overexpression weakened the effects of miR-8485 on cell viability, apoptosis, as well as inflammatory responses. Our data suggest that miR-8485 attenuates the injury of cardiomyocytes by targeting TP53INP1, suggesting it is a protective factor against CHF.
Collapse
Affiliation(s)
- Xiuying Luo
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanlin Xu
- Department of Nephrology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ze Zhong
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng Xiang
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xindong Wu
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Aiguo Chong
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Network-Based Pharmacology and Bioinformatics Study on the Mechanism of Action of Gujiansan in the Treatment of Steroid-Induced Avascular Necrosis of the Femoral Head. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8080679. [PMID: 35915795 PMCID: PMC9338865 DOI: 10.1155/2022/8080679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022]
Abstract
Objective To investigate the main pharmacological basis and mechanism of action of Gujiansan in the treatment of steroid-induced avascular necrosis of the femoral head (SANFH). Methods The active constituents and targets of Gujiansan were screened by using TCMSP and other databases, and relevant disease targets were obtained by analyzing the microarray of SANFH in the GEO database. The intersection of the two was taken to obtain the potential targets of Gujiansan for the treatment of SANFH, and key active constituents were screened with the “active constituent-target” network constructed by the Cytoscape software; then, the STRING database was used to construct the protein interaction network to screen the key targets. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses of key targets were performed by the DAVID database, and the relationship between the “key active constituent-key target-key signaling pathway” was explored. Finally, the molecular docking between key active constituents and key targets was verified. In addition, qPCR detection technology was used to evaluate the preventive and therapeutic effects of key active constituents of Gujiansan in a rat osteoblast model of SANFH to verify the possible mechanism of the effect of Gujiansan in the treatment of SANFH. Results (1) 106 active constituents and 55 targets were obtained for the treatment of SANFH. (2) Quercetin, luteolin, kaempferol, cryptotanshinone, and naringenin were the key active constituents for the treatment of SANFH. (3) IL1B, STAT3, CAT, PTGS2, and MAPK3 were the key targets for the treatment of SANFH. (4) IL1B, STAT3, CAT, PTGS2, MAPK3, and HMOX1 are key targets in the protein interaction network. (5) DAVID enrichment analysis mainly covers the regulation of DNA-binding transcription factor activity, positive regulation of cytokine production, and response to oxidative stress and other biological processes, involving IL-17, AGE-RAGE, C-type lectin receptor, and other signaling pathways. (6) Gujiansan is a multitarget and multisignaling pathway for the treatment of SANFH. (7) Good binding activity exists between key active constituents and key targets. Conclusion This study analyzes the potential mechanism of action of Gujiansan in the treatment of SANFH with network pharmacology, which can provide a reference for the further study of its pharmacological basis and targets.
Collapse
|
36
|
PRDX2 Knockdown Inhibits Extracellular Matrix Synthesis of Chondrocytes by Inhibiting Wnt5a/YAP1/CTGF and Activating IL-6/JAK2/STAT3 Pathways in Deer Antler. Int J Mol Sci 2022; 23:ijms23095232. [PMID: 35563622 PMCID: PMC9103832 DOI: 10.3390/ijms23095232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 02/05/2023] Open
Abstract
Although peroxiredoxin 2 (PRDX2) plays a vital role in relieving oxidative stress, its physiological function in cartilage development remains almost unknown. In this study, we found that the expression of PRDX2 significantly increased in the chondrocytes compared with pre-chondrocytes. PRDX2 knockdown significantly decreased the expression of extracellular matrix (ECM) protein (Col2a and Aggrecan), which led to blocked cartilage formation. Moreover, PRDX2 knockdown also inhibited the expression of connective tissue growth factor (CTGF). CTGF is an important growth factor that regulates synthesis of ECM proteins. We explored the possible regulatory mechanism by which PRDX2 regulated the expression of CTGF. Our results demonstrated that PRDX2 knockdown downregulated the expression of CTGF by inhibiting Wnt5a/Yes-associated protein 1 (YAP1) pathway. In addition, PRDX2 knockdown promoted the expression of interleukin 6 (IL-6), indicating PRDX2 expression had an anti-inflammatory function during antler growth. Mechanistically, PRDX2 knockdown promoted cartilage matrix degradation by activating the IL-6-mediated Janus Kinase 2/Signal Transducer and Activator of Transcription 3 (JAK2/STAT3) signaling pathway. These results reveal that PRDX2 is a potential regulator that promotes cartilage extracellular matrix synthesis.
Collapse
|
37
|
Shang X, Fang Y, Xin W, You H. The Application of Extracellular Vesicles Mediated miRNAs in Osteoarthritis: Current Knowledge and Perspective. J Inflamm Res 2022; 15:2583-2599. [PMID: 35479833 PMCID: PMC9037713 DOI: 10.2147/jir.s359887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/08/2022] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a whole joint disease characterized by synovitis, cartilage destruction, and subchondral bone sclerosis and cyst. Despite decades’ study, effective treatment is rare for this chronic disease. Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptosis bodies, are nano-sized vesicles with a cargo containing biologically active agents, such as nucleic acids, lipids, and proteins. As a group of short non-coding RNAs, microRNAs (miRNAs) can be delivered by parental cells secreted EVs. Negatively regulate the target mRNAs at the posttranscriptional level and regulate gene expression in recipient cells without modifying gene sequence. Recently, most studies focused on the function of EVs mediated miRNAs in the pathophysiological process of OA. However, all kinds of EVs specific and OA specific factors might influence the administration of EVs-miRNAs, especially the precise quantitative management. As a result, the flourishing of current research about EVs in the laboratory might not promote the relevant clinical transformation in OA treatment. In this review, we reviewed the present application of EVs-miRNAs in the therapeutic of OA and further analyzed the potential factors that might influence its application. Further progress in the quantitative management of EVs-miRNAs would accelerate the clinical transformation of miRNAs enriched EVs in the OA field.
Collapse
Affiliation(s)
- Xiaobin Shang
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yan Fang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Wenqiang Xin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 352000, People’s Republic of China
| | - Hongbo You
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Correspondence: Hongbo You, Email
| |
Collapse
|
38
|
Xian Bo S, Chen W, Chang L, Hao Ran Y, Hui Hui G, Ya Kun Z, Wu Kun X, Hai Tao F, Wen Dan C. The Research Progress of Exosomes in Osteoarthritis, With Particular Emphasis on the Therapeutic Effect. Front Pharmacol 2022; 13:731756. [PMID: 35308214 PMCID: PMC8924513 DOI: 10.3389/fphar.2022.731756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Exosomes participate in many physiological and pathological processes by regulating cell-to-cell communication. This affects the etiology and development of diseases, such as osteoarthritis (OA). Although exosomes in the OA tissue microenvironment are involved in the progression of OA, exosomes derived from therapeutic cells represent a new therapeutic strategy for OA treatment. Recent studies have shown that exosomes participate in OA treatment by regulating the proliferation, apoptosis, inflammation, and extracellular matrix synthesis of chondrocytes. However, studies in this field are scant. This review summarizes the therapeutic properties of exosomes on chondrocytes in OA and their underlying molecular mechanisms. We also discuss the challenges and prospects of exosome-based OA treatment.
Collapse
Affiliation(s)
- Shang Xian Bo
- Anhui Medical University, Hefei, China.,Second Hospital of Anhui Medical University, Hefei, China
| | - Wang Chen
- Anhui Medical University, Hefei, China.,Second Hospital of Anhui Medical University, Hefei, China
| | - Liu Chang
- Armed Police Corps Hospital of Anhui Province, Hefei, China
| | - Yu Hao Ran
- Second Hospital of Anhui Medical University, Hefei, China
| | - Guo Hui Hui
- Anhui Medical University, Hefei, China.,Second Hospital of Anhui Medical University, Hefei, China
| | - Zhu Ya Kun
- Fuyang Hospital of Anhui Medical University, Anhui, China
| | - Xie Wu Kun
- Second Hospital of Anhui Medical University, Hefei, China
| | - Fan Hai Tao
- Fuyang Hospital of Anhui Medical University, Anhui, China
| | - Cheng Wen Dan
- Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
39
|
Yan C, Yu J. Noncoding RNA in Extracellular Vesicles Regulate Differentiation of Mesenchymal Stem Cells. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2021.806001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To achieve the desired outcome in tissue engineering regeneration, mesenchymal stem cells need to undergo a series of biological processes, including differentiating into the ideal target cells. The extracellular vesicle (EV) in the microenvironment contributes toward determining the fate of the cells with epigenetic regulation, particularly from noncoding RNA (ncRNA), and exerts transportation and protective effects on ncRNAs. We focused on the components and functions of ncRNA (particularly microRNA) in the EVs. The EVs modified by the ncRNA favor tissue regeneration and pose a potential challenge.
Collapse
|
40
|
Exosomes in the Pathogenesis, Progression, and Treatment of Osteoarthritis. Bioengineering (Basel) 2022; 9:bioengineering9030099. [PMID: 35324788 PMCID: PMC8945849 DOI: 10.3390/bioengineering9030099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a prevalent and debilitating age-related joint disease characterized by articular cartilage degeneration, synovial membrane inflammation, osteophyte formation, as well as subchondral bone sclerosis. OA drugs at present are mainly palliative and do not halt or reverse disease progression. Currently, no disease-modifying OA drugs (DMOADs) are available and total joint arthroplasty remains a last resort. Therefore, there is an urgent need for the development of efficacious treatments for OA management. Among all novel pharmaco-therapeutical options, exosome-based therapeutic strategies are highly promising. Exosome cargoes, which include proteins, lipids, cytokines, and various RNA subtypes, are potentially capable of regulating intercellular communications and gene expression in target cells and tissues involved in OA development. With extensive research in recent years, exosomes in OA studies are no longer limited to classic, mesenchymal stem cell (MSC)-derived vesicles. New origins, structures, and functions of exosomes are constantly being discovered and investigated. This review systematically summarizes the non-classic origins, biosynthesis, and extraction of exosomes, describes modification and delivery techniques, explores their role in OA pathogenesis and progression, and discusses their therapeutic potential and hurdles to overcome in OA treatment.
Collapse
|
41
|
Amsar RM, Wijaya CH, Ana ID, Hidajah AC, Notobroto HB, Kencana Wungu TD, Barlian A. Extracellular vesicles: a promising cell-free therapy for cartilage repair. Future Sci OA 2022; 8:FSO774. [PMID: 35070356 PMCID: PMC8765097 DOI: 10.2144/fsoa-2021-0096] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/12/2021] [Indexed: 11/23/2022] Open
Abstract
Few effective therapies for cartilage repair have been found as cartilage has a low regenerative capacity. Extracellular vesicles (EVs), including exosomes, are produced by cells and contain bioactive components such as nucleic acids, proteins, lipids and other metabolites that have potential for treating cartilage injuries. Challenges like the difficulty in standardizing targeted therapy have prevented EVs from being used frequently as a treatment option. In this review we present current studies, mechanisms and delivery strategies of EVs. Additionally, we describe the challenges and future directions of EVs as therapeutic agents for cartilage repair. Repairing cartilage damage is challenging due to the tissue’s low regenerative capacity. Extracellular vesicles (EVs) contain bioactive components that may be able to treat cartilage injuries. However, EV-based therapy is not widely used. This review summarizes the current state of knowledge regarding the use of EVs for cartilage repair, including the mechanisms, delivery strategies, challenges and future directions.
Collapse
Affiliation(s)
- Rizka Musdalifah Amsar
- School of Life Science & Technology, Institut Teknologi Bandung, Bandung, West Java, 40132, Indonesia
| | - Christofora Hanny Wijaya
- Department of Food Science & Technology, Bogor Agricultural University, West Java, 16680, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Gadjah Mada University, Yogyakarta, 55281, Indonesia
| | - Atik Choirul Hidajah
- Department of Epidemiology Faculty of Public Health, Airlangga University, East Java, 60115, Indonesia
| | - Hari Basuki Notobroto
- Department of Biostatics & Population Faculty of Public Health, Airlangga University, East Java, 60115, Indonesia
| | - Triati Dewi Kencana Wungu
- Nuclear Physics & Biophysics Research Group, Department of Physics, Faculty of Mathematics & Natural Sciences, Institut Teknologi Bandung, West Java, 40132, Indonesia
- Research Center for Nanoscience & Nanotechnology, Institut Teknologi Bandung, West Java, 40132, Indonesia
| | - Anggraini Barlian
- School of Life Science & Technology, Institut Teknologi Bandung, Bandung, West Java, 40132, Indonesia
- Research Center for Nanoscience & Nanotechnology, Institut Teknologi Bandung, West Java, 40132, Indonesia
| |
Collapse
|
42
|
Scrapie-Responsive Gene 1 Promotes Chondrogenic Differentiation of Umbilical Cord Mesenchymal Stem Cells via Wnt5a. Stem Cells Int 2022; 2022:9124277. [PMID: 35126528 PMCID: PMC8813292 DOI: 10.1155/2022/9124277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/25/2021] [Accepted: 12/29/2021] [Indexed: 11/17/2022] Open
Abstract
Objective Repair of cartilage defects, a common condition resulting from many factors, is still a great challenge. Based on their chondrogenic differentiation ability, mesenchymal stem cell- (MSC-) based cartilage regeneration is a promising approach for cartilage defect repair. However, MSC differentiation into chondroblasts or related cell lineages is elaborately controlled by stem cell differentiation stage factors and affected by an array of bioactive elements, which may impede the efficient production of target cells. Thus, identifying a single transcription factor to promote chondrogenic differentiation is critical. Herein, we explored the mechanism by which scrapie-responsive gene 1 (SCRG1), a candidate gene for cartilage regeneration promotion, regulates chondrogenic differentiation of MSCs. Methods Expression of SCRG1 was detected in umbilical cord-derived MSCs (UCMSCs) by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemical analysis during chondrogenic differentiation. The function of SCRG1 in chondrogenic potential was evaluated after gene knockdown or overexpression by lentiviral vectors. Finally, a rabbit cartilage defect model was established to evaluate the effect of SCRG1 on cartilage repair in vivo. Results Expression of SCRG1 was upregulated during in vitro chondrogenic differentiation of UCMSCs. SCRG1 knockdown inhibited chondrogenic differentiation of UCMSCs, while SCRG1 overexpression promoted chondrogenic differentiation of UCMSCs in vitro. In addition, UCMSC overexpressing SCRG1 promoted cartilage repair in vivo. Mechanistically, SCRG1 promoted chondrogenic differentiation via upregulation of Wnt5a expression and subsequent inhibition of β-catenin. Conclusion Our results showed that SCRG1 promotes chondrogenic differentiation of UCMSCs by inhibiting canonical Wnt/β-catenin signaling through Wnt5a. Our findings provide a future target for chondrogenic differentiation and cartilage regeneration.
Collapse
|
43
|
Li H, Zheng Q, Xie X, Wang J, Zhu H, Hu H, He H, Lu Q. Role of Exosomal Non-Coding RNAs in Bone-Related Diseases. Front Cell Dev Biol 2022; 9:811666. [PMID: 35004702 PMCID: PMC8733689 DOI: 10.3389/fcell.2021.811666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Bone-related diseases seriously affect the lives of patients and carry a heavy economic burden on society. Treatment methods cannot meet the diverse clinical needs of affected patients. Exosomes participate in the occurrence and development of many diseases through intercellular communication, including bone-related diseases. Studies have shown that exosomes can take-up and “package” non-coding RNAs and “deliver” them to recipient cells, thereby regulating the function of recipient cells. The exosomal non-coding RNAs secreted by osteoblasts, osteoclasts, chondrocytes, and other cells are involved in the regulation of bone-related diseases by inhibiting osteoclasts, enhancing chondrocyte activity and promoting angiogenesis. Here, we summarize the role and therapeutic potential of exosomal non-coding RNAs in the bone-related diseases osteoporosis, osteoarthritis, and bone-fracture healing, and discuss the clinical application of exosomes in patients with bone-related diseases.
Collapse
Affiliation(s)
- Hang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Qiyue Zheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyan Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China.,College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Haihong Zhu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Haoye Hu
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hao He
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
44
|
Yin B, Ni J, Witherel CE, Yang M, Burdick JA, Wen C, Wong SHD. Harnessing Tissue-derived Extracellular Vesicles for Osteoarthritis Theranostics. Theranostics 2022; 12:207-231. [PMID: 34987642 PMCID: PMC8690930 DOI: 10.7150/thno.62708] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a prevalent chronic whole-joint disease characterized by low-grade systemic inflammation, degeneration of joint-related tissues such as articular cartilage, and alteration of bone structures that can eventually lead to disability. Emerging evidence has indicated that synovium or articular cartilage-secreted extracellular vesicles (EVs) contribute to OA pathogenesis and physiology, including transporting and enhancing the production of inflammatory mediators and cartilage degrading proteinases. Bioactive components of EVs are known to play a role in OA include microRNA, long non-coding RNA, and proteins. Thus, OA tissues-derived EVs can be used in combination with advanced nanomaterial-based biosensors for the diagnostic assessment of OA progression. Alternatively, mesenchymal stem cell- or platelet-rich plasma-derived EVs (MSC-EVs or PRP-EVs) have high therapeutic value for treating OA, such as suppressing the inflammatory immune microenvironment, which is often enriched by pro-inflammatory immune cells and cytokines that reduce chondrocytes apoptosis. Moreover, those EVs can be modified or incorporated into biomaterials for enhanced targeting and prolonged retention to treat OA effectively. In this review, we explore recently reported OA-related pathological biomarkers from OA joint tissue-derived EVs and discuss the possibility of current biosensors for detecting EVs and EV-related OA biomarkers. We summarize the applications of MSC-EVs and PRP-EVs and discuss their limitations for cartilage regeneration and alleviating OA symptoms. Additionally, we identify advanced therapeutic strategies, including engineered EVs and applying biomaterials to increase the efficacy of EV-based OA therapies. Finally, we provide our perspective on the future of EV-related diagnosis and therapeutic potential for OA treatment.
Collapse
Affiliation(s)
- Bohan Yin
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Junguo Ni
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | | | - Mo Yang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, PA 16802, USA.,✉ Corresponding authors: Jason A. Burdick: . Chunyi Wen: . Siu Hong Dexter Wong:
| | - Chunyi Wen
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China.,Research Institute of Smart Ageing, the Hong Kong Polytechnic University, Hong Kong, 999077, China.,✉ Corresponding authors: Jason A. Burdick: . Chunyi Wen: . Siu Hong Dexter Wong:
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China.,✉ Corresponding authors: Jason A. Burdick: . Chunyi Wen: . Siu Hong Dexter Wong:
| |
Collapse
|
45
|
Chen J, Yu X, Zhang X. Advances on biological functions of exosomal non-coding RNAs in osteoarthritis. Cell Biochem Funct 2021; 40:49-59. [PMID: 34921424 DOI: 10.1002/cbf.3679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/24/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023]
Abstract
Exosomes can be secreted by various cells and function as intercellular communication vehicles by delivering specific cargoes from the donor cells to the recipient cells through their paracrine activity. Recently, an increasing number of studies have shown that non-coding RNAs (ncRNAs) could be entrapped in and transferred between cartilage-related cells as exosomal cargoes to modulate the expression of various target genes by regulation at post-transcriptional and post-translational levels. They are mainly comprised of microRNAs, long non-coding RNAs, and circular RNAs. Articular cartilage degeneration is one of the main pathological features of osteoarthritis. Exosomal ncRNAs are involved in pathological processes of osteoarthritis, such as proliferation, migration, chondrogenesis, chondrocyte differentiation induction, extracellular matrix formation, apoptosis, and inflammation. In this review, we summarize the biological functions of exosomal ncRNAs in cartilage homeostasis and osteoarthritis progression and discuss the perspectives and challenges of exosomal ncRNAs application for osteoarthritis patients in the future. Exosomal ncRNA has an important regulatory role in the pathogenesis of osteoarthritis, but more evidence is needed for clinical application.
Collapse
Affiliation(s)
- Jialei Chen
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Yu
- Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Tang Z, Lu Y, Zhang S, Wang J, Wang Q, Xiao Y, Zhang X. Chondrocyte secretome enriched microparticles encapsulated with the chondrocyte membrane to facilitate the chondrogenesis of BMSCs and reduce hypertrophy. J Mater Chem B 2021; 9:9989-10002. [PMID: 34874033 DOI: 10.1039/d1tb02319e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Co-culture of chondrocytes and mesenchymal stem cells (MSCs) represents an effective way to stimulate the chondrogenesis of MSCs and reduce hypertrophy, but the limited donor site supply and the requirement of two-stage operations are among the major barriers of using autologous chondrocytes in clinical settings. With recent evidence indicating that the chondrogenic effects of the above co-culture mainly lied on the paracrine secretion, and that cell membranes also played crucial roles during the chondrocyte-MSC interaction, we fabricated a multifunctional design of "artificial chondrocytes", which consist of chondrocyte secretome enriched PLGA microparticles with the encapsulation of chondrocytes' membrane fragments. The artificial chondrocytes had shown a similar diameter and surface electrical charge to natural chondrocytes, with the preserved key chondrocyte membrane surface proteins and sustainedly released chondrogenic cytokines from the chondrocyte secretome to extend their effects in vivo. Consequently, the co-culture studies of artificial chondrocytes and bone marrow MSCs had shown the beneficial effects from both chondrocyte secretome and membrane fragments, which also synergistically facilitated the cell proliferation, chondrogenic gene expression, cartilaginous matrix production, and reduced phenotypic hypertrophy in vitro and in vivo. Together, this study has successfully developed the proof-of-concept design of "artificial chondrocytes", which could potentially conquer many major barriers of using natural chondrocytes and provided a novel synthetic-cell approach to current therapeutical strategies towards the functional regeneration of articular cartilage.
Collapse
Affiliation(s)
- Zizhao Tang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Yan Lu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Shixin Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Jing Wang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu, Sichuan, 610059, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China. .,College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610065, China
| |
Collapse
|
47
|
Static Magnetic Fields Enhance the Chondrogenesis of Mandibular Bone Marrow Mesenchymal Stem Cells in Coculture Systems. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9962861. [PMID: 34873576 PMCID: PMC8643226 DOI: 10.1155/2021/9962861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/20/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022]
Abstract
Objectives Combining the advantages of static magnetic fields (SMF) and coculture systems, we investigated the effect of moderate-intensity SMF on the chondrogenesis and proliferation of mandibular bone marrow mesenchymal stem cells (MBMSCs) in the MBMSC/mandibular condylar chondrocyte (MCC) coculture system. The main aim of the present study was to provide an experimental basis for obtaining better cartilage tissue engineering seed cells for the effective repair of condylar cartilage defects in clinical practice. Methods MBMSCs and MCCs were isolated from SD (Sprague Dawley) rats. Flow cytometry, three-lineage differentiation, colony-forming assays, immunocytochemistry, and toluidine blue staining were used for the identification of MBMSCs and MCCs. MBMSCs and MCCs were seeded into the lower and upper Transwell chambers, respectively, at a ratio of 1 : 2, and exposed to a 280 mT SMF. MBMSCs were harvested after 3, 7, or 14 days for analysis. CCK-8 was used to detect cell proliferation, Alcian blue staining was utilized to evaluate glycosaminoglycan (GAG), and western blotting and real-time quantitative polymerase chain reaction (RT-qPCR) detected protein and gene expression levels of SOX9, Col2A1 (Collagen Type II Alpha 1), and Aggrecan (ACAN). Results The proliferation of MBMSCs was significantly enhanced in the experimental group with MBMSCs cocultured with MCCs under SMF stimulation relative to controls (P < 0.05). GAG content was increased, and SOX9, Col2A1, and ACAN were also increased at the mRNA and protein levels (P < 0.05). Conclusions Moderate-intensity SMF improved the chondrogenesis and proliferation of MBMSCs in the coculture system, and it might be a promising approach to repair condylar cartilage defects in the clinical setting.
Collapse
|
48
|
Schizas NP, Zafeiris C, Neri AA, Anastasopoulos PP, Papaioannou NA, Dontas IA. Inhibition versus activation of canonical Wnt-signaling, to promote chondrogenic differentiation of Mesenchymal Stem Cells. A review. Orthop Rev (Pavia) 2021; 13:27098. [PMID: 34745485 DOI: 10.52965/001c.27098] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 12/31/2022] Open
Abstract
Canonical Wnt signaling regulation is essential for controlling stemness and differentiation of mesenchymal stem cells (MSCs). However, the mechanism through which canonical Wnt-dependent MSC lineage commitment leads to chondrogenesis is controversial. Some studies hypothesize that inhibition of canonical Wnt signaling induces MSC chondrogenic differentiation, while others support that the pathway should be activated to achieve MSC chondrogenesis. The purpose of the present review is to analyze data from recent studies to elucidate parameters regarding the role of canonical Wnt signaling in MSC chondrogenic differentiation.
Collapse
Affiliation(s)
| | | | | | | | | | - Ismene A Dontas
- School of Medicine, National and Kapodistrian University of Athens
| |
Collapse
|
49
|
Research Progress on the Antiosteoarthritic Mechanism of Action of Natural Products. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7714533. [PMID: 34630617 PMCID: PMC8497106 DOI: 10.1155/2021/7714533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022]
Abstract
Background Osteoarthritis (OA) is a clinical joint degenerative disease, the pathogenic factors of which include age, obesity, and mechanical injury. Its main pathological features include cartilage loss, narrowing of joint space, and osteophyte formation. At present, there are a variety of treatment methods for OA. Natural products, which are gradually being applied in the treatment of OA, are advantageous as they present with low toxicity and low costs and act on multiple targets. Methods The terms “natural products,” “osteoarthritis,” and “chondrocytes” were searched in PubMed to screen the related literature in the recent 10 years. Results We comprehensively introduced 62 published papers on 48 natural products involving 6, 3, 5, 12, 4, and 5 kinds of terpenoids, polysaccharides, polyphenols, flavonoids, alkaloids, and saponins, respectively (and others). Conclusion The mechanisms of their anti-OA action mainly involve reducing the production of inflammatory factors, reducing oxidative stress, regulating the metabolism of chondrocytes, promoting the proliferation of chondrocytes, or inhibiting chondrocyte apoptosis. This article summarizes the anti-OA activity of natural products in the last 10 years and provides candidate monomers for further study for use in OA treatment.
Collapse
|
50
|
Li Z, Xue H, Tan G, Xu Z. Effects of miRNAs, lncRNAs and circRNAs on osteoporosis as regulatory factors of bone homeostasis (Review). Mol Med Rep 2021; 24:788. [PMID: 34505632 PMCID: PMC8441966 DOI: 10.3892/mmr.2021.12428] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/25/2021] [Indexed: 01/03/2023] Open
Abstract
Osteoporosis is a common metabolic bone disorder typically characterized by decreased bone mass and an increased risk of fracture. At present, the detailed molecular mechanism underlying the development of osteoporosis remains to be elucidated. Accumulating evidence shows that non-coding (nc)RNAs, such as microRNAs (miRNAs), long ncRNAs (lncRNAs) and circular RNAs (circRNAs), play significant roles in osteoporosis through the post-transcriptional regulation of gene expression as regulatory factors. Previous studies have demonstrated that ncRNAs participate in maintaining bone homeostasis by regulating physiological and developmental processes in osteoblasts, osteoclasts and bone marrow stromal cells. In the present review, the latest research investigating the involvement of miRNAs, lncRNAs and circRNAs in regulating the differentiation, proliferation, apoptosis and autophagy of cells that maintain the bone microenvironment in osteoporosis is summarized. Deeper insight into the aspects of osteoporosis pathogenesis involving the deregulation of ncRNAs could facilitate the development of therapeutic approaches for osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Haipeng Xue
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Guoqing Tan
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Zhanwang Xu
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| |
Collapse
|