1
|
Han Z, Wang K, Ding S, Zhang M. Cross-talk of inflammation and cellular senescence: a new insight into the occurrence and progression of osteoarthritis. Bone Res 2024; 12:69. [PMID: 39627227 PMCID: PMC11615234 DOI: 10.1038/s41413-024-00375-z] [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: 01/31/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 12/06/2024] Open
Abstract
Osteoarthritis (OA) poses a significant challenge in orthopedics. Inflammatory pathways are regarded as central mechanisms in the onset and progression of OA. Growing evidence suggests that senescence acts as a mediator in inflammation-induced OA. Given the lack of effective treatments for OA, there is an urgent need for a clearer understanding of its pathogenesis. In this review, we systematically summarize the cross-talk between cellular senescence and inflammation in OA. We begin by focusing on the mechanisms and hallmarks of cellular senescence, summarizing evidence that supports the relationship between cellular senescence and inflammation. We then discuss the mechanisms of interaction between cellular senescence and inflammation, including senescence-associated secretory phenotypes (SASP) and the effects of pro- and anti-inflammatory interventions on cellular senescence. Additionally, we focus on various types of cellular senescence in OA, including senescence in cartilage, subchondral bone, synovium, infrapatellar fat pad, stem cells, and immune cells, elucidating their mechanisms and impacts on OA. Finally, we highlight the potential of therapies targeting senescent cells in OA as a strategy for promoting cartilage regeneration.
Collapse
Affiliation(s)
- Zeyu Han
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China
| | - Ketao Wang
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China
| | - Shenglong Ding
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China
| | - Mingzhu Zhang
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China.
| |
Collapse
|
2
|
Steens W, Zinser W, Rößler P, Heyse T. Infiltration therapy in the context of cartilage surgery. Arch Orthop Trauma Surg 2024; 144:3913-3923. [PMID: 37400671 PMCID: PMC11564373 DOI: 10.1007/s00402-023-04964-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
Guideline-based surgical cartilage therapy for focal cartilage damage offers highly effective possibilities to sustainably reduce patients' complaints and to prevent or at least delay the development of early osteoarthritis. In the knee joint, it has the potential to reduce almost a quarter of the arthroses requiring joint replacement caused by cartilage damage. Biologically effective injection therapies could further improve these results. Based on the currently available literature and preclinical studies, intra- and postoperative injectables may have a positive effect of platelet-rich plasma/fibrin (PRP/PRF) and hyaluronic acid (HA) on cartilage regeneration and, in the case of HA injections, also on the clinical outcome can be assumed. The role of a combination therapy with use of intra-articular corticosteroids is lacking in the absence of adequate study data and cannot be defined yet. With regard to adipose tissue-based cell therapy, the current scientific data do not yet justify any recommendation for its use. Further studies also regarding application intervals, timing and differences in different joints are required.
Collapse
Affiliation(s)
- Wolfram Steens
- Department of Orthopaedics, University Medicine, 18057, Rostock, Germany.
- Orthopaedic-Neurosurgery Center, Roentgenstrasse 10, 45661, Recklinghausen, Germany.
| | - Wolfgang Zinser
- Orthoexpert, 8724, Knittelfeld, Austria
- AUVA-Unfallkrankenhaus Steiermark, 8775, Kalwang, Austria
| | - Philip Rößler
- Joint Center, Middelrhine, 56068, Koblenz, Germany
- Department of Orthopaedic and Trauma Surgery, University Hospital Bonn, 53127, Bonn, Germany
| | - Thomas Heyse
- Center of Orthopedics and Traumatology, University Hospital Marburg, 35033, Marburg, Germany
- Orthomedic Joint Center, Frankfurt Offenbach, 63065, Offenbach, Germany
| |
Collapse
|
3
|
Zhang Y, Chen J, Sun Y, Wang M, Liu H, Zhang W. Endogenous Tissue Engineering for Chondral and Osteochondral Regeneration: Strategies and Mechanisms. ACS Biomater Sci Eng 2024; 10:4716-4739. [PMID: 39091217 DOI: 10.1021/acsbiomaterials.4c00603] [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: 08/04/2024]
Abstract
Increasing attention has been paid to the development of effective strategies for articular cartilage (AC) and osteochondral (OC) regeneration due to their limited self-reparative capacities and the shortage of timely and appropriate clinical treatments. Traditional cell-dependent tissue engineering faces various challenges such as restricted cell sources, phenotypic alterations, and immune rejection. In contrast, endogenous tissue engineering represents a promising alternative, leveraging acellular biomaterials to guide endogenous cells to the injury site and stimulate their intrinsic regenerative potential. This review provides a comprehensive overview of recent advancements in endogenous tissue engineering strategies for AC and OC regeneration, with a focus on the tissue engineering triad comprising endogenous stem/progenitor cells (ESPCs), scaffolds, and biomolecules. Multiple types of ESPCs present within the AC and OC microenvironment, including bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived mesenchymal stem cells (AD-MSCs), synovial membrane-derived mesenchymal stem cells (SM-MSCs), and AC-derived stem/progenitor cells (CSPCs), exhibit the ability to migrate toward injury sites and demonstrate pro-regenerative properties. The fabrication and characteristics of scaffolds in various formats including hydrogels, porous sponges, electrospun fibers, particles, films, multilayer scaffolds, bioceramics, and bioglass, highlighting their suitability for AC and OC repair, are systemically summarized. Furthermore, the review emphasizes the pivotal role of biomolecules in facilitating ESPCs migration, adhesion, chondrogenesis, osteogenesis, as well as regulating inflammation, aging, and hypertrophy-critical processes for endogenous AC and OC regeneration. Insights into the applications of endogenous tissue engineering strategies for in vivo AC and OC regeneration are provided along with a discussion on future perspectives to enhance regenerative outcomes.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
4
|
Feng K, Wang F, Chen H, Zhang R, Liu J, Li X, Xie X, Kang Q. Cartilage progenitor cells derived extracellular vesicles-based cell-free strategy for osteoarthritis treatment by efficient inflammation inhibition and extracellular matrix homeostasis restoration. J Nanobiotechnology 2024; 22:345. [PMID: 38890638 PMCID: PMC11186174 DOI: 10.1186/s12951-024-02632-z] [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: 12/30/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease which currently lacks of effective agents. It is therefore urgent and necessary to seek an effective approach that can inhibit inflammation and promote cartilage matrix homeostasis. Cartilage progenitor cells (CPCs) are identified as a cell population of superficial zone in articular cartilage which possess strong migration ability, proliferative capacity, and chondrogenic potential. Recently, the application of CPCs may represent a novel cell therapy strategy for OA treatment. There is growing evidence that extracellular vesicles (EVs) are primary mediators of the benefits of stem cell-based therapy. In this study, we explored the protective effects of CPCs-derived EVs (CPCs-EVs) on IL-1β-induced chondrocytes. We found CPCs-EVs exhibited chondro-protective effects in vitro. Furthermore, our study demonstrated that CPCs-EVs promoted matrix anabolism and inhibited inflammatory response at least partially via blocking STAT3 activation. In addition, liquid chromatography-tandem mass spectrometry analysis identified 991 proteins encapsulated in CPCs-EVs. By bioinformatics analysis, we showed that STAT3 regulatory proteins were enriched in CPCs-EVs and could be transported to chondrocytes. To promoting the protective function of CPCs-EVs in vivo, CPCs-EVs were modified with cationic peptide ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) for surface charge reverse. In posttraumatic OA mice, our results showed PPD modified CPCs-EVs (PPD-EVs) effectively inhibited extracellular matrix catabolism and attenuated cartilage degeneration. Moreover, PPD-EVs down-regulated inflammatory factors expressions and reduced OA-related pain in OA mice. In ex-vivo cultured OA cartilage explants, PPD-EVs successfully promoted matrix anabolism and inhibited inflammation. Collectively, CPCs-EVs-based cell-free therapy is a promising strategy for OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Feng Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Hongfang Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Rui Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiashuo Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaodong Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xuetao Xie
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
5
|
Iseki T, Rothrauff BB, Kihara S, Overholt KJ, Taha T, Lin H, Alexander PG, Tuan RS. Enhanced osteochondral repair by leukocyte-depleted platelet-rich plasma in combination with adipose-derived mesenchymal stromal cells encapsulated in a three-dimensional photocrosslinked injectable hydrogel in a rabbit model. Stem Cell Res Ther 2024; 15:159. [PMID: 38831361 PMCID: PMC11149275 DOI: 10.1186/s13287-024-03750-z] [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: 07/11/2023] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
INTRODUCTION Intra-articular injection of adipose-derived mesenchymal stromal cells (ASCs) and/or platelet-rich plasma (PRP) have been reported to independently and synergistically improve healing of osteochondral lesions in animal models. However, their independent and combined effects when localized to an osteochondral lesion by encapsulation within a photocrosslinkable methacrylated gelatin hydrogel (GelMA) have not been explored. Herein we investigated a unique combination of allogeneic ASCs and PRP embedded in GelMA as a single-stage treatment for osteochondral regeneration in a rabbit model. METHODS Thirty mature rabbits were divided into six experimental groups: (1) Sham; (2) Defect; (3) GelMA; (4) GelMA + ASCs; (5) GelMA + PRP; and (6) GelMA + ASCs + PRP.At 12 weeks following surgical repair, osteochondral regeneration was assessed on the basis of gross appearance, biomechanical properties, histological and immunohistochemical characteristics, and subchondral bone volume. RESULTS In terms of mechanical property reflecting the ability of neotissue to bear stress, PRP only group were significantly lower than the Sham group (p = 0.0098). On the other hand, ASCs only and ASCs combined with PRP groups did not exhibit significantly difference, which suggesting that incorporation of ASCs assists in restoring the ability of the neotissue to bear stresses similarly to native tissue (p = 0.346, p = 0.40, respectively). Safranin O in ASCs combined with PRP group was significantly higher than the Defect and GelMA only groups (p = 0.0009, p = 0.0017, respectively). Additionally, ASCs only and ASCs combined with PRP groups presented especially strong staining for collagen type II. Surprisingly, PRP only and PRP + ASCs groups tended to exhibit higher collagen type I and collagen type X staining compared to ASCs only group, suggesting a potential PRP-mediated hypertrophic effect. CONCLUSION Regeneration of a focal osteochondral defect in a rabbit model was improved by a single-stage treatment of a photocrosslinked hydrogel containing allogenic ASCs and autologous PRP, with the combination of ASCs and PRP producing superior benefit than either alone. No experimental construct fully restored all properties of the native, healthy osteochondral unit, which may require longer follow-up or further modification of PRP and/or ASCs characteristics.
Collapse
Affiliation(s)
- Tomoya Iseki
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- Department of Orthopaedic Surgery, Hyogo Medical University, 1-1 Mukogawa-cho, 663-8501, Nishinomiya City, Hyogo, Japan
| | - Benjamin B Rothrauff
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Shinsuke Kihara
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Kalon J Overholt
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Tarek Taha
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
6
|
Atwal A, Dale TP, Snow M, Forsyth NR, Davoodi P. Injectable hydrogels: An emerging therapeutic strategy for cartilage regeneration. Adv Colloid Interface Sci 2023; 321:103030. [PMID: 37907031 DOI: 10.1016/j.cis.2023.103030] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
The impairment of articular cartilage due to traumatic incidents or osteoarthritis has posed significant challenges for healthcare practitioners, researchers, and individuals suffering from these conditions. Due to the absence of an approved treatment strategy for the complete restoration of cartilage defects to their native state, the tissue condition often deteriorates over time, leading to osteoarthritic (OA). However, recent advancements in the field of regenerative medicine have unveiled promising prospects through the utilization of injectable hydrogels. This versatile class of biomaterials, characterized by their ability to emulate the characteristics of native articular cartilage, offers the distinct advantage of minimally invasive administration directly to the site of damage. These hydrogels can also serve as ideal delivery vehicles for a diverse range of bioactive agents, including growth factors, anti-inflammatory drugs, steroids, and cells. The controlled release of such biologically active molecules from hydrogel scaffolds can accelerate cartilage healing, stimulate chondrogenesis, and modulate the inflammatory microenvironment to halt osteoarthritic progression. The present review aims to describe the methods used to design injectable hydrogels, expound upon their applications as delivery vehicles of biologically active molecules, and provide an update on recent advances in leveraging these delivery systems to foster articular cartilage regeneration.
Collapse
Affiliation(s)
- Arjan Atwal
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Tina P Dale
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Martyn Snow
- Department of Arthroscopy, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham B31 2AP, United Kingdom; The Robert Jones and Agnes Hunt Hospital, Oswestry, Shropshire SY10 7AG, United Kingdom
| | - Nicholas R Forsyth
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom; Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Pooya Davoodi
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom.
| |
Collapse
|
7
|
Li ZL, Li XT, Hao RC, Wang FY, Wang YX, Zhao ZD, Li PL, Yin BF, Mao N, Ding L, Zhu H. Human osteoarthritic articular cartilage stem cells suppress osteoclasts and improve subchondral bone remodeling in experimental knee osteoarthritis partially by releasing TNFAIP3. Stem Cell Res Ther 2023; 14:253. [PMID: 37752608 PMCID: PMC10523665 DOI: 10.1186/s13287-023-03411-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 07/07/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Though articular cartilage stem cell (ACSC)-based therapies have been demonstrated to be a promising option in the treatment of diseased joints, the wide variety of cell isolation, the unknown therapeutic targets, and the incomplete understanding of the interactions of ACSCs with diseased microenvironments have limited the applications of ACSCs. METHODS In this study, the human ACSCs have been isolated from osteoarthritic articular cartilage by advantage of selection of anatomical location, the migratory property of the cells, and the combination of traumatic injury, mechanical stimuli and enzymatic digestion. The protective effects of ACSC infusion into osteoarthritis (OA) rat knees on osteochondral tissues were evaluated using micro-CT and pathological analyses. Moreover, the regulation of ACSCs on osteoarthritic osteoclasts and the underlying mechanisms in vivo and in vitro were explored by RNA-sequencing, pathological analyses and functional gain and loss experiments. The one-way ANOVA was used in multiple group data analysis. RESULTS The ACSCs showed typical stem cell-like characteristics including colony formation and committed osteo-chondrogenic capacity. In addition, intra-articular injection into knee joints yielded significant improvement on the abnormal subchondral bone remodeling of osteoarthritic rats. Bioinformatic and functional analysis showed that ACSCs suppressed osteoarthritic osteoclasts formation, and inflammatory joint microenvironment augmented the inhibitory effects. Further explorations demonstrated that ACSC-derived tumor necrosis factor alpha-induced protein 3 (TNFAIP3) remarkably contributed to the inhibition on osteoarhtritic osteoclasts and the improvement of abnormal subchondral bone remodeling. CONCLUSION In summary, we have reported an easy and reproducible human ACSC isolation strategy and revealed their effects on subchondral bone remodeling in OA rats by releasing TNFAIP3 and suppressing osteoclasts in a diseased microenvironment responsive manner.
Collapse
Affiliation(s)
- Zhi-Ling Li
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
| | - Xiao-Tong Li
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
| | - Rui-Cong Hao
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
- Basic Medical College of Anhui Medical University, Hefei, 230032, Anhui Province, People's Republic of China
| | - Fei-Yan Wang
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
- Basic Medical College of Anhui Medical University, Hefei, 230032, Anhui Province, People's Republic of China
| | - Yu-Xing Wang
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, People's Republic of China
| | - Zhi-Dong Zhao
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, People's Republic of China
| | - Pei-Lin Li
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
| | - Bo-Feng Yin
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China
| | - Ning Mao
- Beijing Institute of Basic Medical Sciences, Road Taiping 27, Beijing, 100850, People's Republic of China
| | - Li Ding
- Air Force Medical Center, PLA, Road Fucheng 30, Beijing, 100142, People's Republic of China.
| | - Heng Zhu
- Department of Stem Cells and Regenerative Medicine, Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.
- Basic Medical College of Anhui Medical University, Hefei, 230032, Anhui Province, People's Republic of China.
| |
Collapse
|
8
|
Huang Y, Zhao H, Wang Y, Bi S, Zhou K, Li H, Zhou C, Wang Y, Wu W, Peng B, Tang J, Pan B, Wang B, Chen Z, Li Z, Zhang Z. The application and progress of tissue engineering and biomaterial scaffolds for total auricular reconstruction in microtia. Front Bioeng Biotechnol 2023; 11:1089031. [PMID: 37811379 PMCID: PMC10556751 DOI: 10.3389/fbioe.2023.1089031] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/21/2023] [Indexed: 10/10/2023] Open
Abstract
Microtia is a congenital deformity of the ear with an incidence of about 0.8-4.2 per 10,000 births. Total auricular reconstruction is the preferred treatment of microtia at present, and one of the core technologies is the preparation of cartilage scaffolds. Autologous costal cartilage is recognized as the best material source for constructing scaffold platforms. However, costal cartilage harvest can lead to donor-site injuries such as pneumothorax, postoperative pain, chest wall scar and deformity. Therefore, with the need of alternative to autologous cartilage, in vitro and in vivo studies of biomaterial scaffolds and cartilage tissue engineering have gradually become novel research hot points in auricular reconstruction research. Tissue-engineered cartilage possesses obvious advantages including non-rejection, minimally invasive or non-invasive, the potential of large-scale production to ensure sufficient donors and controllable morphology. Exploration and advancements of tissue-engineered cartilaginous framework are also emerging in aspects including three-dimensional biomaterial scaffolds, acquisition of seed cells and chondrocytes, 3D printing techniques, inducing factors for chondrogenesis and so on, which has greatly promoted the research process of biomaterial substitute. This review discussed the development, current application and research progress of cartilage tissue engineering in auricular reconstruction, particularly the usage and creation of biomaterial scaffolds. The development and selection of various types of seed cells and inducing factors to stimulate chondrogenic differentiation in auricular cartilage were also highlighted. There are still confronted challenges before the clinical application becomes widely available for patients, and its long-term effect remains to be evaluated. We hope to provide guidance for future research directions of biomaterials as an alternative to autologous cartilage in ear reconstruction, and finally benefit the transformation and clinical application of cartilage tissue engineering and biomaterials in microtia treatment.
Collapse
Affiliation(s)
- Yeqian Huang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hanxing Zhao
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Yixi Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siwei Bi
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Zhou
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Hairui Li
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Yudong Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqing Wu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Peng
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Jun Tang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Bo Pan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoyun Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhixing Chen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Wen Y, Chen Y, Wu W, Zhang H, Peng Z, Yao X, Zhang X, Jiang W, Liao Y, Xie Y, Shen X, Sun H, Hu J, Liu H, Chen X, Chen J, Ouyang H. Hyperplastic Human Macromass Cartilage for Joint Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301833. [PMID: 37395375 PMCID: PMC10502860 DOI: 10.1002/advs.202301833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/07/2023] [Indexed: 07/04/2023]
Abstract
Cartilage damage affects millions of people worldwide. Tissue engineering strategies hold the promise to provide off-the-shelf cartilage analogs for tissue transplantation in cartilage repair. However, current strategies hardly generate sufficient grafts, as tissues cannot maintain size growth and cartilaginous phenotypes simultaneously. Herein, a step-wise strategy is developed for fabricating expandable human macromass cartilage (macro-cartilage) in a 3D condition by employing human polydactyly chondrocytes and a screen-defined serum-free customized culture (CC). CC-induced chondrocytes demonstrate improved cell plasticity, expressing chondrogenic biomarkers after a 14.59-times expansion. Crucially, CC-chondrocytes form large-size cartilage tissues with average diameters of 3.25 ± 0.05 mm, exhibiting abundant homogenous matrix and intact structure without a necrotic core. Compared with typical culture, the cell yield in CC increases 2.57 times, and the expression of cartilage marker collagen type II increases 4.70 times. Transcriptomics reveal that this step-wise culture drives a proliferation-to-differentiation process through an intermediate plastic stage, and CC-chondrocytes undergo a chondral lineage-specific differentiation with an activated metabolism. Animal studies show that CC macro-cartilage maintains a hyaline-like cartilage phenotype in vivo and significantly promotes the healing of large cartilage defects. Overall, an efficient expansion of human macro-cartilage with superior regenerative plasticity is achieved, providing a promising strategy for joint regeneration.
Collapse
|
10
|
Zhang C, Wang G, An Y. Achieving Nasal Septal Cartilage In Situ Regeneration: Focus on Cartilage Progenitor Cells. Biomolecules 2023; 13:1302. [PMID: 37759702 PMCID: PMC10527213 DOI: 10.3390/biom13091302] [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: 06/23/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
The nasal septal cartilage plays an important role in preventing the collapse of the nasal bones and maintaining the appearance of the nose. In the context of inherent difficulties regarding septal cartilage repair and the shortage of cartilage graft resources for regeneration, tissue engineering, especially the in situ strategy based on scaffolds, has become a new prospect and become one of the most promising approaches. Given that it is difficult for chondrocytes to achieve directional migration and secrete matrix components to participate in tissue repair after cartilage injury, cartilage progenitor cells (CPCs), with great migratory ability and stem cell characteristics, have caught the attention of researchers and brought hope for nasal septal cartilage in situ regeneration. In this review, we first summarized the distribution, characteristics, isolation, and culture methods of nasal septal CPCs. Subsequently, we described the roles of migratory CPCs in cartilage regeneration. Finally, we reviewed the existing studies on CPCs-based cartilage tissue engineering and summarized the strategies for promoting the migration and chondrogenesis of CPCs so as to provide ideas for achieving nasal septal cartilage in situ regeneration.
Collapse
Affiliation(s)
| | | | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; (C.Z.)
| |
Collapse
|
11
|
Chen Y, Liao G, Ma T, Li L, Yang J, Shen B, Lu Y, Si H. YY1/miR-140-5p/Jagged1/Notch axis mediates cartilage progenitor/stem cells fate reprogramming in knee osteoarthritis. Int Immunopharmacol 2023; 121:110438. [PMID: 37295026 DOI: 10.1016/j.intimp.2023.110438] [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: 04/05/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/11/2023]
Abstract
Osteoarthritis is a multifactorial disease characterized by cartilage degeneration, while cartilage progenitor/stem cells (CPCs) are responsible for endogenous cartilage repair. However, the relevant regulatory mechanisms of CPCs fate reprogramming in OA are rarely reported. Recently, we observed fate disorders in OA CPCs and found that microRNA-140-5p (miR-140-5p) protects CPCs from fate changes in OA. This study further mechanistically investigated the upstream regulator and downstream effectors of miR-140-5p in OA CPCs fate reprogramming. As a result, luciferase reporter assay and validation assays revealed that miR-140-5p targets Jagged1 and inhibits Notch signaling in human CPCs, and the loss-/gain-of-function experiments and rescue assays discovered that miR-140-5p improves OA CPCs fate, but this effect can be counteracted by Jagged1. Moreover, increased transcription factor Ying Yang 1 (YY1) was associated with OA progression, and YY1 could disturb CPCs fate via transcriptionally repressing miR-140-5p and enhancing the Jagged1/Notch signaling. Finally, the relevant changes and mechanisms of YY1, miR-140-5p, and Jagged1/Notch signaling in OA CPCs fate reprogramming were validated in rats. Conclusively, this study identified a novel YY1/miR-140-5p/Jagged1/Notch signaling axis that mediates OA CPCs fate reprogramming, wherein YY1 and Jagged1/Notch signaling exhibits an OA-stimulative role, and miR-140-5p plays an OA-protective effect, providing attractive targets for OA therapeutics.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guangneng Liao
- Experimental Animal Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Ma
- Department of Operating Room of Anesthesia Surgery Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lan Li
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Yang
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Shen
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanrong Lu
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haibo Si
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
12
|
Feng K, Yu Y, Chen Z, Wang F, Zhang K, Chen H, Xu J, Kang Q. Injectable hypoxia-preconditioned cartilage progenitor cells-laden GelMA microspheres system for enhanced osteoarthritis treatment. Mater Today Bio 2023; 20:100637. [PMID: 37128287 PMCID: PMC10148185 DOI: 10.1016/j.mtbio.2023.100637] [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: 12/29/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 05/03/2023] Open
Abstract
Osteoarthritis (OA) is the most common age-related degenerative joint disease mainly characterized by the destruction of articular cartilage. Owing to its native avascular property, intrinsic repair of articular cartilage is very limited. Thus, a chondrogenic microenvironment in the joint is essential to the preservation of healthy chondrocytes and OA treatment. Recently, cartilage progenitor cells (CPCs)-based therapy is emerging as a promising strategy to repair degenerated and damaged articular cartilage. In this study, injectable hypoxia-preconditioned three-dimensional (3D) cultured CPCs-laden gelatin methacryloyl (GelMA) microspheres (CGMs) were constructed and characterized. Compared to normoxia-pretreated 3D CPCs and two-dimensional (2D) cultured CPCs, hypoxia-preconditioned 3D cultured CPCs exhibited enhanced cartilage extracellular matrix (ECM) secretion and greater chondrogenic ability. In addition, hypoxia-preconditioned 3D cultured CPCs more effectively maintained cartilage matrix metabolism balance and attenuated articular cartilage degeneration in subacute and chronic rat OA models. Mechanistically, our results demonstrated hypoxia-preconditioned 3D cultured CPCs exerted chondro-protective effects by inhibiting inflammation and oxidative stress via NRF2/HO-1 pathway in vitro and in vivo. Together, through the 3D culture of CPCs using GelMA microspheres (GMs) under hypoxia environment, our results proposed an efficient articular cartilage regeneration strategy for OA treatment and could provide inspiration for other stem cells-based therapies.
Collapse
Affiliation(s)
- Kai Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yifan Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhengsheng Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Feng Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Kunqi Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Hongfang Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
13
|
Vinod E, Lisha J J, Parasuraman G, Livingston A, Daniel AJ, Sathishkumar S. Evaluation of ghrelin as a distinguishing marker for human articular cartilage-derived chondrocytes and chondroprogenitors. J Clin Orthop Trauma 2023; 41:102175. [PMID: 37303495 PMCID: PMC10248861 DOI: 10.1016/j.jcot.2023.102175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/22/2023] [Accepted: 05/28/2023] [Indexed: 06/13/2023] Open
Abstract
Purpose of the study Cell-based therapeutics for articular cartilage repair primarily employed bone marrow-derived mesenchymal stem cells and chondrocytes. Research to overcome their limitation of formation of a functionally poor fibro-hyaline type of repair tissue led to the discovery of chondroprogenitors (CPCs), cartilage resident stem cells. These cells isolated by adhesion assay using fibronectin (FAA-CPs) and migration of progenitors from explants (MCPs) display higher chondrogenic and lower terminal differentiation potential. During in-vitro culture, chondrocytes tend to de-differentiate and acquire characteristics similar to stem cells, thus making it challenging to distinguish them from other cell groups. Ghrelin, a cytoplasmic growth hormone secretagogue, has been proposed to play a vital role in chondrogenesis, with reports of its higher expression in chondrocytes than BM-MSCs. The aim of this study was to compare the mRNA expression of Ghrelin between BM-MSCs, chondrocytes, FAA-CPs and MCP and the possibility of it serving as a distinguishing marker. Methods The four populations isolated from three human osteoarthritic knee joints were characterised by CD marker expression for positive (CD 90, CD73 and CD105) and negative (HLA-DR, CD34 and CD45) MSC markers and trilineage differentiation (adipogenic, osteogenic and chondrogenic) and subjected to qRT-PCR to assess Ghrelin's gene expression. Results This study showed that all groups exhibited similar expression of CD markers and multilineage potential. Though chondrocytes showed greater expression of Ghrelin, it was not statistically significant to classify it as a distinguishing marker between these cell populations. Conclusion Ghrelin does not serve to differentiate the subpopulations in terms of their mRNA expression. Further evaluation using their associated enzymes and receptors could provide valuable information to uncover their potential as unequivocal biomarkers.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Jeya Lisha J
- Department of Physiology, Christian Medical College, Vellore, India
| | - Ganesh Parasuraman
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Abel Livingston
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | - Alfred Job Daniel
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | | |
Collapse
|
14
|
Zhao X, Chen C, Luo Y, Li D, Wang Q, Fang Y, Kang P. Connexin43 overexpression promotes bone regeneration by osteogenesis and angiogenesis in rat glucocorticoid-induced osteonecrosis of the femoral head. Dev Biol 2023; 496:73-86. [PMID: 36805498 DOI: 10.1016/j.ydbio.2023.02.004] [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: 06/29/2022] [Revised: 01/30/2023] [Accepted: 02/12/2023] [Indexed: 02/21/2023]
Abstract
Glucocorticoids induced osteonecrosis of the femoral head (GIONFH) is a devastating orthopedic disease. Previous studies suggested that connexin43 is involved in the process of osteogenesis and angiogenesis. However, the role of Cx43 potentiates in the osteogenesis and angiogenesis of bone marrow-derived stromal stem cells (BMSCs) in GIONFH is still not investigated. In this study, BMSCs were isolated and transfected with green fluorescent protein or the fusion gene encoding GFP and Cx43. The osteogenic differentiation of BMSCs were detected after transfected with Cx43. In addition, the migration abilities and angiogenesis of human umbilical vein endothelial cells (HUVECs) were been detected after induced by transfected BMSCs supernatants in vitro. Finally, we established GC-ONFH rat model, then, a certain amount of transfected or controlled BMSCs were injected into the tibia of the rats. Immunohistological staining and micro-CT scanning results showed that the transplanted experiment group had significantly promoted more bone regeneration and vessel volume when compared with the effects of the negative or control groups. This study demonstrated for the first time that the Cx43 overexpression in BMSCs could promote bone regeneration as seen in the osteogenesis and angiogenesis process, suggesting that Cx43 may serve as a therapeutic gene target for GIONFH treatment.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China; Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Changjun Chen
- Department of Orthopedics, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Yue Luo
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Donghai Li
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Qiuru Wang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Yuying Fang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, Shandong, People's Republic of China.
| | - Pengde Kang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
15
|
Vinod E, Padmaja K, Ramasamy B, Sathishkumar S. Systematic review of articular cartilage derived chondroprogenitors for cartilage repair in animal models. J Orthop 2023; 35:43-53. [PMID: 36387762 PMCID: PMC9647330 DOI: 10.1016/j.jor.2022.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
Purpose of research The potential for cartilage repair using articular cartilage derived chondroprogenitors has recently gained popularity due to promising results from in-vitro and in-vivo studies. Translation of results from in-vitro to a clinical setting requires a sufficient number of animal studies displaying significant positive outcomes. Thus, this systematic review comprehensively discusses the available literature (January 2000-March 2022) on animal models employing chondroprogenitors for cartilage regeneration, highlighting the results and limitations associated with their use.As per Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a web-based search of PubMed and SCOPUS databases was performed for the following terminologies: "chondroprogenitors", "cartilage-progenitors", and "chondrogenic-progenitors", which yielded 528 studies. A total of 12 studies met the standardized inclusion criteria, which included chondroprogenitors derived from hyaline cartilage isolated using fibronectin adhesion assay (FAA) or migratory assay from explant cultures, further analyzing the role of chondroprogenitors using in-vivo animal models. Principal results Analysis revealed that FAA chondroprogenitors demonstrated the ability to attenuate osteoarthritis, repair chondral defects and form stable cartilage in animal models. They displayed better outcomes than bone marrow-derived mesenchymal stem cells but were comparable to chondrocytes. Migratory chondroprogenitors also demonstrated superiority to BM-MSCs in terms of higher chondrogenesis and lower hypertrophy, although a direct comparison to FAA-CPs and other cell types is warranted. Major conclusions Chondroprogenitors exhibit superior properties for chondrogenic repair; however, limited data on animal studies necessitates further studies to optimize their use before clinical translation for neo-cartilage formation.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India
- Centre for Stem Cell Research, (A unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Kawin Padmaja
- Department of Physiology, Christian Medical College, Vellore, India
| | - Boopalan Ramasamy
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | | |
Collapse
|
16
|
Zhao Z, Wang Y, Yin B, Li X, Hao R, Li Z, Li P, Han M, Ding L, Li Z, Zhu H. Defect-adaptive Stem-cell-microcarrier Construct Promotes Tissue Repair in Rabbits with Knee Cartilage Defects. Stem Cell Rev Rep 2023; 19:201-212. [PMID: 35900693 DOI: 10.1007/s12015-022-10421-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 01/29/2023]
Abstract
Although various reconstruction techniques are available for cartilage defects, the repair effects and conveniences remain to be further improved due to the limited regenerative capacity of cartilaginous tissues and difficulties in seamlessly fulfilling irregularly shaped defects. In the current study, we explored the repair efficacy of stem cell microcarrier construct (microcarriers loaded with human chondrogenic progenitor cells or bone marrow mesenchymal stem cells) in cartilage defect models. A total of 39 healthy New Zealand white rabbits were included, and femoral trochlear cartilage defect models were established (n = 33). Stem cell microcarrier constructs were implanted into cartilage defects (n = 6), the maintenance conditions of the implanted constructs were observed on days 4, 8, and 30 post implantation (n = 3). Gross observation and pathological analysis were performed to assay the reconstitution of cartilage defects at 12 weeks post-cartilage defect repair(n = 6). The microcarriers could fill the defect model with good plasticity to integrate well with the boundary native normal cartilage. At 3 months after implantation, the defects were filled with fibrous cartilage tissues in the microcarrier without stem cells group. In the microcarrier loaded with BMSCs group, newly formed tissue with a similar appearance of boundary cartilage fulfilled the defects, but the surface was not completely smooth. Promisingly, the defects were almost completely filled with newly regenerated cartilaginous tissues, which had a smooth appearance similar to that of normal cartilage in the microcarrier loaded with CPCs group. These results suggest the feasibility of stem cell microcarrier construct in repairing cartilage defects, indicating promising clinical application prospects.
Collapse
Affiliation(s)
- Zhidong Zhao
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yuxing Wang
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Bofeng Yin
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Xiaotong Li
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Ruicong Hao
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Graduate School of Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhiling Li
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Peilin Li
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Mengyue Han
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Graduate School of Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li Ding
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China. .,Air Force Medical Center, PLA, No.30 Fucheng Road, Beijing, 100142, China.
| | - Zhongli Li
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China. .,Graduate School of Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
17
|
Chen Y, Huang H, Zhong W, Li L, Lu Y, Si HB. miR-140-5p protects cartilage progenitor/stem cells from fate changes in knee osteoarthritis. Int Immunopharmacol 2023; 114:109576. [PMID: 36527878 DOI: 10.1016/j.intimp.2022.109576] [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: 11/13/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022]
Abstract
Cartilage progenitor/stem cells (CPCs) are promising seed cells for cartilage regeneration, but their fate changes and regulatory mechanisms in osteoarthritis (OA) pathogenesis remain unclear. This study aimed to investigate the role and potential mechanism of the microRNA-140-5p (miR-140-5p), whose protective role in knee OA has been confirmed by our previous studies, in OA CPCs fate reprogramming. Firstly, the normal and OA CPCs were isolated, and the fate indicators, miR-140-5p, Jagged1, and Notch signals were detected and analyzed. Then, the effect of miR-140-5p and the Notch pathway on CPCs fate reprogramming and miR-140-5p on Jagged1/Notch signaling was investigated in IL-1β-induced chondrocytes in vitro. Finally, the effect of miR-140-5p on OA CPCs fate reprogramming and the potential mechanisms were validated in OA rats. As a result, CPCs percentage was increased in the mild OA cartilage-derived total chondrocytes while decreased in the advanced OA group. Significant fate changes (including reduced cell viability, migration, chondrogenesis, and increased apoptosis), increased Jagged1 and Notch signals, and reduced miR-140-5p were observed in OA CPCs and associated with OA progression. IL-1β induced OA-like changes in CPCs fate, which could be exacerbated by miR-140-5p inhibitor while alleviated by DAPT (a specific Notch inhibitor) and miR-140-5p mimic. Finally, the in vitro phenomenal and mechanistic findings were validated in OA rats. Overall, miR-140-5p protects CPCs from fate changes via inhibiting Jagged1/Notch signaling in knee OA, providing attractive targets for OA therapeutics.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hua Huang
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Wen Zhong
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lan Li
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yanrong Lu
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Hai-Bo Si
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
18
|
Qi L, Wang J, Chen X, Ding Y, Ling B, Wang W, Xu J, Xue Z. Single-cell transcriptomics reveals variable trajectories of CSPCs in the progression of osteoarthritis. Heliyon 2022; 8:e11148. [PMID: 36339749 PMCID: PMC9634280 DOI: 10.1016/j.heliyon.2022.e11148] [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: 01/04/2022] [Revised: 05/05/2022] [Accepted: 10/13/2022] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is characterised by cartilage destruction; however, there are no specific drugs available for its treatment. Cartilage-derived stem/progenitor cells (CSPCs) are multipotent cells that play an essential role in cartilage renewal and may provide critical insights into the medical needs for OA treatment. However, alterations in cell function and fate of CSPCs during OA progression have seldom been analysed, especially at the single-cell level. Additionally, it has been reported that CSPCs can migrate to the cartilage injury area, although the mechanism of migration remains elusive. Thus, understanding the changing patterns of CSPCs in the pathological process of OA is important in the effort to develop stem cell therapy for OA. Here, we downloaded single-cell transcriptomic data of patients with OA from the Gene Expression Omnibus (GEO) database and performed unbiased clustering of the cells based on gene expression patterns using the Seurat package. Using common stem cell markers and chondrogenic transcription factors, we traced CSPCs throughout all stages of OA. We further explored the dynamics of CSPCs in OA progression and validated the single-cell RNA sequencing data in vitro using qPCR, immunofluorescence, and western blotting. Specifically, we primarily explored the heterogeneity of CSPCs at the single-cell level and found that it was closely associated with OA progression. Our results indicate significantly reduced chondrogenic differentiation capacity in CSPCs during the late stage of OA, while their proliferation capacity tended to increase. We also found that genes implicated in fibrosis, cell motility, and extracellular matrix remodelling were upregulated in CSPCs during the progression of OA. Our study revealed the dynamics of stem cells in OA progression and may inform the development of stem cell therapy for OA.
Collapse
Affiliation(s)
- Lingbin Qi
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jian Wang
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xian Chen
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanhui Ding
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Bin Ling
- The Second People’s Hospital of Yunnan Province, Kunming, China
| | - Wenjun Wang
- Stem Cell and Regenerative Medicine Engineering Research Center of Hunan Province, Hunan Yuanpin Cell Technology Co. Ltd, 102 Dongwu Road, Changsha City 410100, Hunan Province, China
- Corresponding author.
| | - Jun Xu
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Corresponding author.
| | - Zhigang Xue
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
- Corresponding author.
| |
Collapse
|
19
|
Zhao Z, Li J, Bai X, Wang Y, Wang Q, Lv N, Gao H, Guo Z, Zhu H, Guo Q, Li Z. Microfracture Augmentation With Direct In Situ Radial Shockwave Stimulation With Appropriate Energy Has Comparable Repair Performance With Tissue Engineering in the Porcine Osteochondral Defect Model. Am J Sports Med 2022; 50:3660-3670. [PMID: 36190157 DOI: 10.1177/03635465221125936] [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] [Indexed: 01/31/2023]
Abstract
BACKGROUND The first-line clinical strategy for small cartilage/osteochondral defects is microfracture (MF). However, its repair efficacy needs improvement. HYPOTHESIS Appropriate energy radial shockwave stimulation in MF holes would greatly improve repair efficacy in the porcine osteochondral defect model, and it may obtain comparable performance with common tissue engineering techniques. STUDY DESIGN Controlled laboratory study. METHODS Osteochondral defect models (8-mm diameter, 3-mm depth) were established in the weightbearing area of Bama pigs' medial femoral condyles. In total, 25 minipigs were randomly divided into 5 groups: control (Con; without treatment), MF, MF augmentation (MF+; treated with appropriate energy radial shockwave stimulation in MF holes after MF), tissue engineering (TE; treated with compounds of microcarrier and bone marrow mesenchymal stem cells), and sham (as the positive control). After 3 months of intervention, osteochondral specimens were harvested for macroscopic, radiological, biomechanical, and histological evaluations. The statistical data were analyzed using 1-way analysis of variance. RESULTS Based on the macroscopic appearance, the smoothness and integration of the repaired tissue in the MF+ group were improved when compared with the Con and MF groups. The histological staining suggested more abundant cartilaginous matrix deposition in the MF+ group versus the Con and MF groups. The general scores of the macroscopic and histological appearances were comparable in the MF+ and the TE groups. The high signal areas of the osteochondral unit in the magnetic resonance images were significantly decreased in the MF+ group, with no difference with the TE group. The micro-computed tomography data demonstrated the safety of direct in situ radial shockwave performance. Biomechanical tests revealed that the repaired tissue's Young modulus was highest in the MF+ group and not statistically different from that in the TE group. CONCLUSION Direct in situ radial shockwave stimulation with appropriate energy significantly improves the short-term repair efficacy of MF. More encouragingly, the MF+ group in our study obtained repair performance comparable with the TE therapy. CLINICAL RELEVANCE This strategy is easy to perform and can readily be generalized with safety and higher cartilage repair efficacy. Moreover, it is expected to be accomplished under arthroscopy, indicating tremendous clinical transformative value.
Collapse
Affiliation(s)
- Zhidong Zhao
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ji Li
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaowei Bai
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuxing Wang
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Qi Wang
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ningyu Lv
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Huayi Gao
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zheng Guo
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Quanyi Guo
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhongli Li
- Department of Orthopedics, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
20
|
Infiltration nach chirurgischer Knorpeltherapie. ARTHROSKOPIE 2022. [DOI: 10.1007/s00142-022-00561-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Wu MJM, Sermer C, Kandel RA, Theodoropoulos JS. Characterization of Migratory Cells From Bioengineered Bovine Cartilage in a 3D Co-culture Model. Am J Sports Med 2022; 50:3090-3101. [PMID: 35983988 PMCID: PMC9442774 DOI: 10.1177/03635465221113325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 06/03/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Chondrocyte migration in native cartilage is limited and has been implicated as one of the reasons for the poor integration of native implants. Through use of an in vitro integration model, it has previously been shown that cells from bioengineered cartilage can migrate into the native host cartilage during integration. Platelet-rich plasma (PRP) treatment further enhanced integration of bioengineered cartilage to native cartilage in vitro. However, it is not known how PRP treatment of the bioengineered construct promotes this. HYPOTHESIS PRP supports cell migration from bioengineered cartilage and these migratory cells have the ability to accumulate cartilage-like matrix. STUDY DESIGN Controlled laboratory study. METHODS Osteochondral-like constructs were generated by culturing primary bovine chondrocytes on the top surface of a porous bone substitute biomaterial composed of calcium polyphosphate. After 1 week in culture, the constructs were submerged in PRP and placed adjacent, but 2 mm distant, to a native bovine osteochondral plug in a co-culture model for 2 weeks. Cell migration was monitored using phase-contrast imaging. Cell phenotype was determined by evaluating the gene expression of matrix metalloprotease 13 (MMP-13), Ki67, and cartilage matrix molecules using quantitative polymerase chain reaction. When tissue formed, it was assessed by histology, immunohistochemistry, and quantification of matrix content. RESULTS PRP treatment resulted in the formation of a fiber network connecting the bioengineered cartilage and native osteochondral plug. Cells from both the bioengineered cartilage and the native osteochondral tissue migrated onto the PRP fibers and formed a tissue bridge after 2 weeks of culture. Migratory cells on the tissue bridge expressed higher levels of collagen types II and I (COL2, COL1), Ki67 and MMP-13 mRNA compared with nonmigratory cells in the bioengineered cartilage. Ki67 and MMP-13-positive cells were found on the edges of the tissue bridge. The tissue bridge accumulated COL1 and COL2 and aggrecan and contained comparable collagen and glycosaminoglycan content to the bioengineered cartilage matrix. The tissue bridge did not reliably develop in the absence of cells from the native osteochondral plug. CONCLUSION Bioengineered cartilage formed by bovine chondrocytes contains cells that can migrate on PRP fibers and form cartilaginous tissue. CLINICAL RELEVANCE Migratory cells from bioengineered cartilage may promote cartilage integration. Further studies are required to determine the role of migratory cells in integration in vivo.
Collapse
Affiliation(s)
- Ming Jia Michael Wu
- Lunenfeld-Tanenbaum Research Institute,
Toronto, Ontario, Canada
- Institute of Biomaterials and
Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Corey Sermer
- Lunenfeld-Tanenbaum Research Institute,
Toronto, Ontario, Canada
- Institute of Biomaterials and
Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Rita A. Kandel
- Lunenfeld-Tanenbaum Research Institute,
Toronto, Ontario, Canada
- Institute of Biomaterials and
Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Pathology and Laboratory Medicine,
Mount Sinai Hospital, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology,
University of Toronto, Toronto, Ontario, Canada
| | - John S. Theodoropoulos
- Division of Orthopaedic Surgery, Mount
Sinai Hospital, Toronto, Ontario, Canada
- Division of Orthopaedic Surgery,
University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Wu S, Guo W, Li R, Zhang X, Qu W. Progress of Platelet Derivatives for Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:907356. [PMID: 35782516 PMCID: PMC9243565 DOI: 10.3389/fbioe.2022.907356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Articular cartilage has limited self-regeneration ability for lacking of blood vessels, nerves, and lymph that makes it a great challenge to repair defects of the tissue and restore motor functions of the injured or aging population. Platelet derivatives, such as platelet-rich plasma, have been proved effective, safe, and economical in musculoskeletal diseases for their autologous origin and rich in growth factors. The combination of platelet derivatives with biomaterials provides both mechanical support and localized sustained release of bioactive molecules in cartilage tissue engineering and low-cost efficient approaches of potential treatment. In this review, we first provide an overview of platelet derivatives and their application in clinical and experimental therapies, and then we further discuss the techniques of the addition of platelet derivatives and their influences on scaffold properties. Advances in cartilage tissue engineering with platelet derivatives as signal factors and structural components are also introduced before prospects and concerns in this research field. In short, platelet derivatives have broad application prospects as an economical and effective enhancement for tissue engineering–based articular cartilage repair.
Collapse
Affiliation(s)
- Siyu Wu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Wenlai Guo
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xi Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| | - Wenrui Qu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| |
Collapse
|
23
|
Yin BF, Li ZL, Yan ZQ, Guo Z, Liang JW, Wang Q, Zhao ZD, Li PL, Hao RC, Han MY, Li XT, Mao N, Ding L, Chen DF, Gao Y, Zhu H. Psoralen alleviates radiation-induced bone injury by rescuing skeletal stem cell stemness through AKT-mediated upregulation of GSK-3β and NRF2. Stem Cell Res Ther 2022; 13:241. [PMID: 35672836 PMCID: PMC9172007 DOI: 10.1186/s13287-022-02911-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Repairing radiation-induced bone injuries remains a significant challenge in the clinic, and few effective medicines are currently available. Psoralen is a principal bioactive component of Cullen corylifolium (L.) Medik and has been reported to have antitumor, anti-inflammatory, and pro-osteogenesis activities. However, less information is available regarding the role of psoralen in the treatment of radiation-induced bone injury. In this study, we explored the modulatory effects of psoralen on skeletal stem cells and their protective effects on radiation-induced bone injuries. METHODS The protective effects of psoralen on radiation-induced osteoporosis and irradiated bone defects were evaluated by microCT and pathological analysis. In addition, the cell proliferation, osteogenesis, and self-renewal of SSCs were explored. Further, the underlying mechanisms of the protective of psoralen were investigated by using RNA sequencing and functional gain and loss experiments in vitro and in vivo. Statistical significance was analyzed using Student's t test. The one-way ANOVA was used in multiple group data analysis. RESULTS Here, we demonstrated that psoralen, a natural herbal extract, mitigated radiation-induced bone injury (irradiation-induced osteoporosis and irradiated bone defects) in mice partially by rescuing the stemness of irradiated skeletal stem cells. Mechanistically, psoralen restored the stemness of skeletal stem cells by alleviating the radiation-induced suppression of AKT/GSK-3β and elevating NRF2 expression in skeletal stem cells. Furthermore, the expression of KEAP1 in skeletal stem cells did not significantly change in the presence of psoralen. Moreover, blockade of NRF2 in vivo partially abolished the promising effects of psoralen in a murine model of irradiation-induced osteoporosis and irradiated bone regeneration. CONCLUSIONS In summary, our findings identified psoralen as a potential medicine to mitigate bone radiation injury. In addition, skeletal stem cells and AKT-GSK-3β and NRF2 may thus represent therapeutic targets for treating radiation-induced bone injury.
Collapse
Affiliation(s)
- Bo-Feng Yin
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Zhi-Ling Li
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Zi-Qiao Yan
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, People's Republic of China
| | - Zheng Guo
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, People's Republic of China.,Medical Center of Air Forces, PLA, Road Fucheng 30, Beijing, 100142, People's Republic of China
| | - Jia-Wu Liang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, People's Republic of China.,Medical Center of Air Forces, PLA, Road Fucheng 30, Beijing, 100142, People's Republic of China
| | - Qian Wang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, People's Republic of China.,Medical Center of Air Forces, PLA, Road Fucheng 30, Beijing, 100142, People's Republic of China
| | - Zhi-Dong Zhao
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, People's Republic of China.,Medical Center of Air Forces, PLA, Road Fucheng 30, Beijing, 100142, People's Republic of China
| | - Pei-Lin Li
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Rui-Cong Hao
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,Graduate School of Anhui Medical University, 81 Meishan Road, Shushan Qu, Hefei, 230032, Anhui, People's Republic of China
| | - Meng-Yue Han
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,Graduate School of Anhui Medical University, 81 Meishan Road, Shushan Qu, Hefei, 230032, Anhui, People's Republic of China
| | - Xiao-Tong Li
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Ning Mao
- Beijing Institute of Basic Medical Sciences, Road Taiping 27, Beijing, 100850, People's Republic of China
| | - Li Ding
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China. .,Medical Center of Air Forces, PLA, Road Fucheng 30, Beijing, 100142, People's Republic of China.
| | - Da-Fu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Eastern Street Xinjiekou 31, Beijing, 100035, China.
| | - Yue Gao
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China.
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, People's Republic of China. .,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China. .,Graduate School of Anhui Medical University, 81 Meishan Road, Shushan Qu, Hefei, 230032, Anhui, People's Republic of China. .,Beijing Institute of Basic Medical Sciences, Road Taiping 27, Beijing, 100850, People's Republic of China.
| |
Collapse
|
24
|
Xia D, Wu J, Zhou F, Wang S, Zhang Z, Zhou P, Xu S. Mapping Thematic Trends and Analysing Hotspots Concerning the Use of Stem Cells for Cartilage Regeneration: A Bibliometric Analysis From 2010 to 2020. Front Pharmacol 2022; 12:737939. [PMID: 35046799 PMCID: PMC8762272 DOI: 10.3389/fphar.2021.737939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/19/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Defects of articular cartilage represent a common condition that usually progresses to osteoarthritis with pain and dysfunction of the joint. Current treatment strategies have yielded limited success in these patients. Stem cells are emerging as a promising option for cartilage regeneration. We aim to summarize the developmental history of stem cells for cartilage regeneration and to analyse the relevant trends and hotspots. Methods: We screened all relevant literature on stem cells for cartilage regeneration from Web of Science during 2010–2020 and analysed the research trends in this field by VOSviewer and CiteSpace. We also summarized previous clinical trials. Results: We screened 1,011 publications. China contributed the largest number of publications (317, 31.36%) and citations (81,376, 48.61%). The United States achieved the highest H-index (39). Shanghai Jiao Tong University had the largest number of publications (34) among all full-time institutions. The Journal of Biomaterials and Stem Cell Research and Therapy published the largest number of studies on stem cells for cartilage regeneration (35). SEKIYA I and YANG F published the majority of articles in this field (14), while TOH WS was cited most frequently (740). Regarding clinical research on stem cells for cartilage regeneration, the keyword “double-blind” emerged in recent years, with an average year of 2018.75. In tissue engineering, the keyword “3D printing” appeared latest, with an average year of 2019.625. In biological studies, the key word “extracellular vesicles” appeared latest, with an average year of 2018.9091. The current research trend indicates that basic research is gradually transforming to tissue engineering. Clinical trials have confirmed the safety and feasibility of stem cells for cartilage regeneration. Conclusion: Multiple scientific methods were employed to reveal productivity, collaborations, and research hotspots related to the use of stem cells for cartilage regeneration. 3D printing, extracellular vesicles, and double-blind clinical trials are research hotspots and are likely to be promising in the near future. Further studies are needed for to improve our understanding of this field, and clinical trials with larger sample sizes and longer follow-up periods are needed for clinical transformation.
Collapse
Affiliation(s)
- Demeng Xia
- Department of Orthopedics, Naval Hospital of Eastern Theater, Zhoushan, China.,Department of Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianghong Wu
- Department of Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Feng Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Jiangsu, China
| | - Sheng Wang
- Department of Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhentao Zhang
- Department of Orthopedics, Naval Medical University, Shanghai, China
| | - Panyu Zhou
- Department of Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shuogui Xu
- Department of Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
25
|
Higher Chondrogenic Potential of Extracellular Vesicles Derived from Mesenchymal Stem Cells Compared to Chondrocytes-EVs In Vitro. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9011548. [PMID: 34938811 PMCID: PMC8687842 DOI: 10.1155/2021/9011548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/09/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
Abstract
The inability of cartilage to self-repair necessitates an effective therapeutic approach to restore damaged tissues. Extracellular vesicles (EVs) are attractive options because of their roles in cellular communication and tissue repair where they regulate the cellular processes of proliferation, differentiation, and recruitment. However, it is a challenge to determine the relevant cell sources for isolation of EVs with high chondrogenic potential. The current study aims to evaluate the chondrogenic potential of EVs derived from chondrocytes (Cho-EV) and mesenchymal stem cells (MSC-EV). The EVs were separately isolated from conditioned media of both rabbit bone marrow MSCs and chondrocyte cultures. The isolated vesicles were assessed in terms of size, morphology, and surface marker expression. The chondrogenic potential of MSCs in the presence of different concentrations of EVs (50, 100, and 150 μg/ml) was evaluated during 21 days, and chondrogenic surface marker expressions were checked by qRT-PCR and histologic assays. The extracted vesicles had a spherical morphology and a size of 44.25 ± 8.89 nm for Cho-EVs and 112.1 ± 10.10 nm for MSC-EVs. Both groups expressed the EV-specific surface markers CD9 and CD81. Higher expression of chondrogenic specified markers, especially collagen type II (COL II), and secretion of glycosaminoglycans (GAGs) and proteoglycans were observed in MSCs treated with 50 and 100 μg/ml MSC-EVs compared to the Cho-EVs. The results from the use of EVs, particularly MSC-EVs, with high chondrogenic ability will provide a basis for developing therapeutic agents for cartilage repair.
Collapse
|
26
|
Vinod E, Johnson NN, Kumar S, Amirtham SM, James JV, Livingston A, Rebekah G, Daniel AJ, Ramasamy B, Sathishkumar S. Migratory chondroprogenitors retain superior intrinsic chondrogenic potential for regenerative cartilage repair as compared to human fibronectin derived chondroprogenitors. Sci Rep 2021; 11:23685. [PMID: 34880351 PMCID: PMC8654938 DOI: 10.1038/s41598-021-03082-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Cell-based therapy for articular hyaline cartilage regeneration predominantly involves the use of mesenchymal stem cells and chondrocytes. However, the regenerated repair tissue is suboptimal due to the formation of mixed hyaline and fibrocartilage, resulting in inferior long-term functional outcomes. Current preclinical research points towards the potential use of cartilage-derived chondroprogenitors as a viable option for cartilage healing. Fibronectin adhesion assay-derived chondroprogenitors (FAA-CP) and migratory chondroprogenitors (MCP) exhibit features suitable for neocartilage formation but are isolated using distinct protocols. In order to assess superiority between the two cell groups, this study was the first attempt to compare human FAA-CPs with MCPs in normoxic and hypoxic culture conditions, investigating their growth characteristics, surface marker profile and trilineage potency. Their chondrogenic potential was assessed using mRNA expression for markers of chondrogenesis and hypertrophy, glycosaminoglycan content (GAG), and histological staining. MCPs displayed lower levels of hypertrophy markers (RUNX2 and COL1A1), with normoxia-MCP exhibiting significantly higher levels of chondrogenic markers (Aggrecan and COL2A1/COL1A1 ratio), thus showing superior potential towards cartilage repair. Upon chondrogenic induction, normoxia-MCPs also showed significantly higher levels of GAG/DNA with stronger staining. Focused research using MCPs is required as they can be suitable contenders for the generation of hyaline-like repair tissue.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, India. .,Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India.
| | | | - Sanjay Kumar
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | | | - Jithu Varghese James
- Department of Diabetes, School of Life Course Sciences, King's College London, London, UK
| | - Abel Livingston
- Department of Orthopaedics, Christian Medical College and Hospital, Vellore, India
| | - Grace Rebekah
- Department of Biostatistics, Christian Medical College, Vellore, India
| | - Alfred Job Daniel
- Department of Orthopaedics, Christian Medical College and Hospital, Vellore, India
| | - Boopalan Ramasamy
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, Australia. .,Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia.
| | | |
Collapse
|
27
|
Vinod E, Parameswaran R, Ramasamy B, Kachroo U. Pondering the Potential of Hyaline Cartilage-Derived Chondroprogenitors for Tissue Regeneration: A Systematic Review. Cartilage 2021; 13:34S-52S. [PMID: 32840123 PMCID: PMC8804774 DOI: 10.1177/1947603520951631] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Chondroprogenitors have recently gained prominence due to promising results seen in in vitro and animal studies as a potential contender in cell-based therapy for cartilage repair. Lack of consensus regarding nomenclature, isolation techniques, and expansion protocols create substantial limitations for translational research, especially given the absence of distinct markers of identification. The objective of this systematic review was to identify and collate information pertaining to hyaline cartilage-derived chondroprogenitors, with regard to their isolation, culture, and outcome measures. DESIGN As per Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a web-based search of Scopus and PubMed databases was performed from January 2000 to May 2020, which yielded 509 studies. A total of 65 studies were identified that met the standardized inclusion criteria which comprised of, but was not limited to, progenitors derived from fibronectin adhesion, migrated subpopulation from explant cultures, and single-cell sorting. RESULT Literature search revealed that progenitors demonstrated inherent chondrogenesis and minimal tendency for hypertrophy. Multiple sources also demonstrated significantly better outcomes that bone marrow-derived mesenchymal stem cells and comparable results to chondrocytes. With regard to progenitor subgroups, collated evidence points to better and consistent outcomes with the use of migratory progenitors when compared to fibronectin adhesion assay-derived progenitors, although a direct comparison between the two cell populations is warranted. CONCLUSION Since chondroprogenitors exhibit favorable properties for cartilage repair, efficient characterization of progenitors is imperative, to complete their phenotypic profile, so as to optimize their use in translational research for neocartilage formation.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian
Medical College, Vellore, India,Centre for Stem Cell Research (a unit of
InStem, Bengaluru), Christian Medical College, Vellore, India,Elizabeth Vinod, Department of Physiology,
Christian Medical College, Vellore 632002, Tamil Nadu, India.
| | | | - Boopalan Ramasamy
- Department of Orthopaedics, Royal Darwin
Hospital, Tiwi, Northern Territory, Australia
| | - Upasana Kachroo
- Department of Physiology, Christian
Medical College, Vellore, India
| |
Collapse
|
28
|
Ding L, Han DM, Zheng XL, Yan HM, Xue M, Liu J, Zhu L, Guo ZK, Mao N, Ning HM, Wang HX, Heng Zhu. Infusion of haploidentical hematopoietic stem cells combined with mesenchymal stem cells for treatment of severe aplastic anemia in adult patients yields curative effects. Cytotherapy 2021; 24:205-212. [PMID: 34799271 DOI: 10.1016/j.jcyt.2021.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/11/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND AIMS Despite the great advances in immunosuppressive therapy for severe aplastic anemia (SAA), most patients are not completely cured. Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) has been recommended as an alternative treatment in adult SAA patients. However, haplo-HSCT presents a higher incidence of graft failure and graft-versus-host disease (GVHD). The authors designed a combination of haplo-HSCT and umbilical cord-derived mesenchymal stem cells (UC-MSCs) for treatment of SAA in adult patients and evaluated its effects. METHODS Adult patients (≥18 years) with SAA (N = 25) were given HLA-haploidentical hematopoietic stem cells (HSCs) combined with UC-MSCs after a conditioning regimen consisting of busulfan, cyclophosphamide, fludarabine and anti-thymocyte globulin and intensive GVHD prophylaxis, including cyclosporine, basiliximab, mycophenolate mofetil and short-term methotrexate. Additionally, the effects of the protocol in adult SSA patients were compared with those observed in juvenile SAA patients (N = 75). RESULTS All patients achieved myeloid engraftment after haplo-HSCT at a median of 16.12 days (range, 11-26). The median time of platelet engraftment was 28.30 days (range, 13-143). The cumulative incidence of grade II acute GVHD (aGVHD) at day +100 was 32.00 ± 0.91%. No one had grade III-IV aGVHD at day +100. The cumulative incidence of total chronic GVHD was 28.00 ± 0.85%. The overall survival was 71.78 ± 9.05% at a median follow-up of 42.08 months (range, 2.67-104). Promisingly, the protocol yielded a similar curative effect in both young and adult SAA patients. CONCLUSIONS The authors' data suggest that co-transplantation of HLA-haploidentical HSCs and UC-MSCs may provide an effective and safe treatment for adult SAA.
Collapse
Affiliation(s)
- Li Ding
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China; Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.
| | - Dong-Mei Han
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China
| | - Xiao-Li Zheng
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China
| | - Hong-Min Yan
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China
| | - Mei Xue
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China
| | - Jing Liu
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China
| | - Ling Zhu
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China
| | - Zi-Kuan Guo
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China; Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China; Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Ning Mao
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Hong-Mei Ning
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China; The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Heng-Xiang Wang
- Air Force Medical Center, People's Liberation Army, Beijing, People's Republic of China
| | - Heng Zhu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China; Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China; Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China; Graduate School of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
29
|
Vinod E, Amirtham SM, Kachroo U, Goyal A, Ozbey O, James JV, Sathishkumar S, Ramasamy B. Articular chondroprogenitors in platelet rich plasma for treatment of osteoarthritis and osteochondral defects in a rabbit knee model. Knee 2021; 30:51-62. [PMID: 33857741 DOI: 10.1016/j.knee.2021.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/22/2020] [Accepted: 03/18/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Articular chondroprogenitors are a promising contender for cartilage repair due to their inherent nature which stands primed for chondrogenesis and minimal hypertrophic preponderance. Platelet rich plasma (PRP) has been extensively used for treating cartilage defects and osteoarthritis (OA), due to its chondro-inductive properties and abundant pool of growth factors. The aim of this study was to assess the efficacy of chondroprogenitors injected with PRP versus PRP alone in the healing of experimentally created early OA and osteochondral defects (OCD) in a rabbit model. METHODS Adult New Zealand White male rabbits were used for cell and PRP isolation. Chondroprogenitors were isolated by fibronectin adhesion assay, labelled with iron oxide, characterized for surface markers, differential potential and expanded. PRP was isolated by double spin centrifugation using a TriCell kit. Study groups included (a) Monosodium iodoacetate induced early OA and (b) critical OCD. Following intervention (test arm: PRP+ chondroprogenitors and control arm: PRP), assessment was performed at 6- and 12-weeks which included histopathological examination and scoring (OARSI and Modified Wakitani score), immunohistochemistry analysis (Collagen type II and X) and synovial fluid S100A12 levels. RESULTS AND CONCLUSION Comparable, evident healing was noticed in both test and control arms when the OA group samples were assessed at both time points. In the OCD group, PRP alone exhibited significantly better results than the test arm, although repair was notable in both interventions. Further evaluation of chondroprogenitors is required to assess their role as a standalone therapy and in combination with PRP to further cartilage regeneration.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore 632002, India; Centre for Stem Cell Research, Christian Medical College, Vellore 632002, India.
| | | | - Upasana Kachroo
- Department of Physiology, Christian Medical College, Vellore 632002, India
| | - Anjali Goyal
- Department of Pathology, Smt NHL Municipal Medical College, Ahmedabad 380006, India.
| | - Ozlem Ozbey
- Department of Histology and Embryology Campus, School of Medicine, Akdeniz University, Antalya 07070, Turkey
| | | | | | - Boopalan Ramasamy
- Department of Orthopaedics, Royal Darwin Hospital, 105 Rocklands Drive, Tiwi, NT 0810, Australia.
| |
Collapse
|
30
|
Li PL, Wang YX, Zhao ZD, Li ZL, Liang JW, Wang Q, Yin BF, Hao RC, Han MY, Ding L, Wu CT, Zhu H. Clinical-grade human dental pulp stem cells suppressed the activation of osteoarthritic macrophages and attenuated cartilaginous damage in a rabbit osteoarthritis model. Stem Cell Res Ther 2021; 12:260. [PMID: 33933140 PMCID: PMC8088312 DOI: 10.1186/s13287-021-02353-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/23/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Although increasing evidence has demonstrated that human dental pulp stem cells (hDPSCs) are efficacious for the clinical treatment of skeletal disorders, the underlying mechanisms remain incompletely understood. Osteoarthritis (OA) is one of the most common degenerative disorders in joints and is characterized by gradual and irreversible cartilaginous tissue damage. Notably, immune factors were newly identified to be closely related to OA development. In this study, we explored the modulatory effects of clinical-grade hDPSCs on osteoarthritic macrophages and their protective effects on cartilaginous tissues in OA joints. METHODS The cell morphology, immunophenotype, and inflammatory factor expression of osteoarthritic macrophages were explored by phase contrast microscope, transmission electron microscopy, immunostaining, flow cytometry, quantitative polymerase chain reaction, and enzyme linked immunosorbent assay, respectively. Additionally, the factors and signaling pathways that suppressed macrophage activation by hDPSCs were determined by enzyme-linked immunosorbent assay and western-blotting. Furthermore, hDPSCs were administered to a rabbit knee OA model via intra-articular injection. Macrophage activation in vivo and cartilaginous tissue damage were also evaluated by pathological analysis. RESULTS We found that hDPSCs markedly inhibited osteoarthritic macrophage activation in vitro. The cell morphology, immunophenotype, and inflammatory factor expression of osteoarthritic macrophages changed into less inflammatory status in the presence of hDPSCs. Mechanistically, we observed that hDPSC-derived hepatocyte growth factor and transforming growth factor β1 mediated the suppressive effects on osteoarthritic macrophages. Moreover, phosphorylation of MAPK pathway proteins contributed to osteoarthritic macrophage activation, and hDPSCs suppressed their activation by partially inactivating those pathways. Most importantly, injected hDPSCs inhibited macrophage activation in osteochondral tissues in a rabbit knee OA model in vivo. Further histological analysis showed that hDPSCs alleviated cartilaginous damage to knee joints. CONCLUSIONS In summary, our findings reveal a novel function for hDPSCs in suppressing osteoarthritic macrophages and suggest that macrophages are efficient cellular targets of hDPSCs for alleviation of cartilaginous damage in OA. hDPSCs treat OA via an osteoarthritic macrophages-dependent mechanisms. hDPSCs suppress the activation of osteoarthritic macrophages in vitro and in vivo and alleviate cartilaginous lesions in OA models.
Collapse
Affiliation(s)
- Pei-Lin Li
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Yu-Xing Wang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Zhi-Dong Zhao
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Zhi-Ling Li
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Jia-Wu Liang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Qian Wang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Bo-Feng Yin
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Rui-Cong Hao
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
- Graduate School of Anhui Medical University, 81 Meishan Road, Shu Shan Qu, Hefei, 230032, Anhui, P. R. China
| | - Meng-Yue Han
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
- Graduate School of Anhui Medical University, 81 Meishan Road, Shu Shan Qu, Hefei, 230032, Anhui, P. R. China
| | - Li Ding
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China.
- Medical Center of Air Forces, PLA, Road Fucheng 30, Beijing, 100142, P. R. China.
| | - Chu-Tse Wu
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China.
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China.
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
- Graduate School of Anhui Medical University, 81 Meishan Road, Shu Shan Qu, Hefei, 230032, Anhui, P. R. China.
| |
Collapse
|
31
|
Enhancing the chondrogenic potential of chondrogenic progenitor cells by deleting RAB5C. iScience 2021; 24:102464. [PMID: 34013174 PMCID: PMC8113995 DOI: 10.1016/j.isci.2021.102464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/24/2021] [Accepted: 04/21/2021] [Indexed: 11/21/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease that affects a large proportion of the elderly population. Chondrogenic progenitor cells (CPCs) reside in late-stage OA cartilage tissue, producing a fibrocartilaginous extracellular matrix; these cells can be manipulated in vitro to deposit proteins of healthy articular cartilage. CPCs are under the control of SOX9 and RUNX2. In our earlier studies, we showed that a knockdown of RUNX2 enhanced the chondrogenic potential of CPCs. Here we demonstrate that CPCs carrying a knockout of RAB5C, a protein involved in endosomal trafficking, exhibited elevated expression of multiple chondrogenic markers, including the SOX trio, and increased COL2 deposition, whereas no changes in COL1 deposition were observed. We report RAB5C as an attractive target for future therapeutic approaches designed to increase the COL2 content in the diseased joint.
Collapse
|
32
|
Qi Y, Tang R, Shi Z, Feng G, Zhang W. Wnt5a/Platelet-rich plasma synergistically inhibits IL-1β-induced inflammatory activity through NF-κB signaling pathway and prevents cartilage damage and promotes meniscus regeneration. J Tissue Eng Regen Med 2021; 15:612-624. [PMID: 33843153 DOI: 10.1002/term.3198] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/30/2020] [Accepted: 04/06/2021] [Indexed: 12/25/2022]
Abstract
Noncanonical Wnt5a is a particularly attractive growth factor to maintain chondrogenesis. Platelet-rich plasma (PRP) is an autologous blood-derived product and a source of bioactive growth factors involved in tissue regeneration. The present study aimed to investigate the effect and inflammation reaction of Wnt5a/PRP on meniscus cells, and evaluate meniscus regeneration and osteoarthritis (OA) prevention by the application of Wnt5a/PRP gel in a rabbit model of massive meniscal defect. In vitro, the proliferation, migration, differentiation, and interleukin-1 beta (IL-1β) IL-1β-induced inflammation reaction of meniscus cells treated by Wnt5a and PRP was assessed. In vivo, the anterior half of the medial meniscus of 18 New Zealand rabbits was excised and implanted with PRP gel, Wnt5a/PRP gel or untreated. After 6 and 12 weeks, the regenerated meniscus were evaluated. Wnt5a can promote the migration of meniscus cells. PRP and Wnt5a had synergistic effect in promoting the proliferation and chondrogenic differentiation of meniscus cells. The IL-1β-induced meniscus cells study showed that PRP and Wnt5a had the anti-inflammatory actions through nuclear factor kB (NF-κB) signaling pathway. PRP and Wnt5a/PRP significantly inhibited the increase of the p-p65/p65 and p-IκB-α/IκB-α ratios. In vivo transplantation of Wnt5a/PRP gel was demonstrated to promote meniscus regeneration, while reducing OA of knee joint. Wnt5a with PRP had the anti-inflammatory activity in an IL-1β-induced inflammatory model. They can synergistically improve the chondorgenic differentiation of meniscus cells. Wnt5a/PRP gel treatment could potentially be developed into a new method for meniscus regeneration and the prevention of OA.
Collapse
Affiliation(s)
- Yiying Qi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruofu Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zuobing Shi
- Department of Orthopedic Surgery, Liangzhu Hospital, Hangzhou, Zhejiang, China
| | - Gang Feng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenkan Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
33
|
Wang K, Li J, Wang Y, Wang Y, Qin Y, Yang F, Zhang M, Zhu H, Li Z. Orchestrated cellular, biochemical, and biomechanical optimizations endow platelet-rich plasma-based engineered cartilage with structural and biomechanical recovery. Bioact Mater 2021; 6:3824-3838. [PMID: 33937588 PMCID: PMC8065202 DOI: 10.1016/j.bioactmat.2021.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
Recently, biomaterials for cartilage regeneration has been intensively investigated. However, the development of scaffolds that capture regenerated cartilage with biomechanical and structural recovery has rarely been reported. To address this challenge, platelet-rich plasma (PRP)-based cartilage constructs with a well-orchestrated symphony of cellular, biochemical and biomechanical elements were prepared by simultaneously employing chondrogenic progenitor cells (CPCs) as a cell source, optimizing platelet concentration, and adding an enzyme-ion activator. It was shown that this triple-optimized PRP + CPC construct possessed increased biomechanical properties and suitable biochemical signals. The following in vitro study demonstrated that the triple-optimized PRP + CPC constructs generated cartilage-like tissue with higher expression levels of chondrogenic-specific markers, more deposition of cartilage-specific extracellular matrix (ECM), and greater biomechanical values than those of the other constructs. Twelve weeks after the construct was implanted in a cartilage defect in vivo, histological analysis, qPCR, and biomechanical tests collectively showed that the triple-optimized constructs yielded a more chondrocyte-like cell phenotype with a higher synthesis of Col-II and aggrecan. More importantly, the triple-optimized constructs facilitated cartilage regeneration with better biomechanical recovery than that of the other constructs. These results demonstrate the efficacy of the triple-optimization strategy and highlight the simplicity and potency of this PRP + CPC construct for cartilage regeneration. Cartilage tissue engineering has been intensively investigated. We designed a PRP-based construct with favorable cell source, reinforced scaffold and appropriate biofactors. This designed construct can facilitate cartilage regeneration with biomechanical and structural recovery simultaneously. The favorable performance of the proposed scaffolds highlights the triple-optimization strategy to improve cartilage engineering.
Collapse
Affiliation(s)
- Ketao Wang
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China.,Department of Foot and Ankle, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.,Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ji Li
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - Yuxing Wang
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - Yaqiang Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuanyuan Qin
- Department of Blood Transfusion, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - Fei Yang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Mingzhu Zhang
- Department of Foot and Ankle, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Heng Zhu
- Beijing Institute of Radiation Medicine/Beijing Institute of Basic Medical Sciences, Haidian, Beijing, 100850, China
| | - Zhongli Li
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| |
Collapse
|
34
|
Liang JW, Li PL, Wang Q, Liao S, Hu W, Zhao ZD, Li ZL, Yin BF, Mao N, Ding L, Zhu H. Ferulic acid promotes bone defect repair after radiation by maintaining the stemness of skeletal stem cells. Stem Cells Transl Med 2021; 10:1217-1231. [PMID: 33750031 PMCID: PMC8284777 DOI: 10.1002/sctm.20-0536] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/02/2021] [Accepted: 02/13/2021] [Indexed: 12/14/2022] Open
Abstract
The reconstruction of irradiated bone defects after settlement of skeletal tumors remains a significant challenge in clinical applications. In this study, we explored radiation‐induced skeletal stem cell (SSC) stemness impairments and rescuing effects of ferulic acid (FA) on SSCs in vitro and in vivo. The immunophenotype, cell renewal, cell proliferation, and differentiation of SSCs in vitro after irradiation were investigated. Mechanistically, the changes in tissue regeneration‐associated gene expression and MAPK pathway activation in irradiated SSCs were evaluated. The regenerative capacity of SSCs in the presence of FA in an irradiated bone defect mouse model was also investigated. We found that irradiation reduced CD140a‐ and CD105‐positive cells in skeletal tissues and mouse‐derived SSCs. Additionally, irradiation suppressed cell proliferation, colony formation, and osteogenic differentiation of SSCs. The RNA‐Seq results showed that tissue regeneration‐associated gene expression decreased, and the Western blotting results demonstrated the suppression of phosphorylated p38/MAPK and ERK/MAPK in irradiated SSCs. Notably, FA significantly rescued the radiation‐induced impairment of SSCs by activating the p38/MAPK and ERK/MAPK pathways. Moreover, the results of imaging and pathological analyses demonstrated that FA enhanced the bone repair effects of SSCs in an irradiated bone defect mouse model substantially. Importantly, inhibition of the p38/MAPK and ERK/MAPK pathways in SSCs by specific chemical inhibitors partially abolished the promotive effect of FA on SSC‐mediated bone regeneration. In summary, our findings reveal a novel function of FA in repairing irradiated bone defects by maintaining SSC stemness and suggest that the p38/MAPK and ERK/MAPK pathways contribute to SSC‐mediated tissue regeneration postradiation.
Collapse
Affiliation(s)
- Jia-Wu Liang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Pei-Lin Li
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Qian Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Song Liao
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Wei Hu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Zhi-Dong Zhao
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Zhi-Ling Li
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Bo-Feng Yin
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Ning Mao
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Li Ding
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Air Force Medical Center, PLA, Beijing, People's Republic of China
| | - Heng Zhu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Experimental Hematology & Biochemistry, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China.,Graduate School of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
35
|
Zhao Z, Wang Y, Wang Q, Liang J, Hu W, Zhao S, Li P, Zhu H, Li Z. Radial extracorporeal shockwave promotes subchondral bone stem/progenitor cell self-renewal by activating YAP/TAZ and facilitates cartilage repair in vivo. Stem Cell Res Ther 2021; 12:19. [PMID: 33413606 PMCID: PMC7792202 DOI: 10.1186/s13287-020-02076-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Radial extracorporeal shockwave (r-ESW), an innovative and noninvasive technique, is gaining increasing attention in regenerative medicine due to its mechanobiological effects. Subchondral bone stem/progenitor cells (SCB-SPCs), originating from the pivotal zone of the osteochondral unit, have been shown to have multipotency and self-renewal properties. However, thus far, little information is available regarding the influences of r-ESW on the biological properties of SCB-SPCs and their therapeutic effects in tissue regeneration. METHODS SCB-SPCs were isolated from human knee plateau osteochondral specimens and treated with gradient doses of r-ESW in a suspension stimulation system. The optimized parameters for SCB-SPC self-renewal were screened out by colony-forming unit fibroblast assay (CFU-F). Then, the effects of r-ESW on the proliferation, apoptosis, and multipotency of SCB-SPCs were evaluated. Moreover, the repair efficiency of radial shockwave-preconditioned SCB-SPCs was evaluated in vivo via an osteochondral defect model. Potential mechanisms were explored by western blotting, confocal laser scanning, and high-throughput sequencing. RESULTS The CFU-F data indicate that r-ESW could augment the self-renewal of SCB-SPCs in a dose-dependent manner. The CCK-8 and flow cytometry results showed that the optimized shockwave markedly promoted SCB-SPC proliferation but had no significant influence on cell apoptosis. Radial shockwave exerted no significant influence on osteogenic capacity but strongly suppressed adipogenic ability in the current study. For chondrogenic potentiality, the treated SCB-SPCs were mildly enhanced, while the change was not significant. Importantly, the macroscopic scores and further histological analysis strongly demonstrated that the in vivo therapeutic effects of SCB-SPCs were markedly improved post r-ESW treatment. Further analysis showed that the cartilage-related markers collagen II and proteoglycan were expressed at higher levels compared to their counterpart group. Mechanistic studies suggested that r-ESW treatment strongly increased the expression of YAP and promoted YAP nuclear translocation in SCB-SPCs. More importantly, self-renewal was partially blocked by the YAP-specific inhibitor verteporfin. Moreover, the high-throughput sequencing data indicated that other self-renewal-associated pathways may also be involved in this process. CONCLUSION We found that r-ESW is capable of promoting the self-renewal of SCB-SPCs in vitro by targeting YAP activity and strengthening its repair efficiency in vivo, indicating promising application prospects.
Collapse
Affiliation(s)
- Zhidong Zhao
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Yuxing Wang
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Qian Wang
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Jiawu Liang
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Wei Hu
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Sen Zhao
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Peilin Li
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China. .,Graduate School of Anhui Medical University, No. 81 Meishan Road, Shu Shan District, Hefei, 230032, Anhui Province, China.
| | - Zhongli Li
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
36
|
Fang J, Wang X, Jiang W, Zhu Y, Hu Y, Zhao Y, Song X, Zhao J, Zhang W, Peng J, Wang Y. Platelet-Rich Plasma Therapy in the Treatment of Diseases Associated with Orthopedic Injuries. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:571-585. [PMID: 32380937 DOI: 10.1089/ten.teb.2019.0292] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Platelet-rich plasma (PRP) is an autologous platelet concentrate prepared from the whole blood that is activated to release growth factors (GFs) and cytokines and has been shown to have the potential capacity to reduce inflammation and improve tissue anabolism for regeneration. The use of PRP provides a potential for repair due to its abundant GFs and cytokines, which are key in initiating and modulating regenerative microenvironments for soft and hard tissues. Among outpatients, orthopedic injuries are common and include bone defects, ligament injury, enthesopathy, musculoskeletal injury, peripheral nerve injury, chronic nonhealing wounds, articular cartilage lesions, and osteoarthritis, which are caused by trauma, sport-related or other types of trauma, or tumor resection. Surgical intervention is often required to treat these injuries. However, for numerous reasons regarding limited regeneration capacity and insufficient blood supply of the defect region, these treatments commonly result in unsatisfactory outcomes, and follow-up treatment is challenging. The aim of the present review is to explore future research in the field of PRP therapy in the treatment of diseases associated with orthopedic injuries. Impact statement In recent years, platelet-rich plasma (PRP) has become widely used in the treatment of diseases associated with orthopedic injuries, and the results of numerous studies are encouraging. Due to diseases associated with orthopedic injuries being common in clinics, as a conservative treatment, more and more doctors and patients are more likely to accept PRP. Importantly, PRP is a biological product of autologous blood that is obtained by a centrifugation procedure to enrich platelets from whole blood, resulting in few complications, such as negligible immunogenicity from an autologous source, and it is also simple to produce through an efficient and cost-effective method in a sterile environment. However, the applicability, advantages, and disadvantages of PRP therapy have not yet been fully elucidated. The aim of the present review is to explore future research in the field of PRP therapy in the treatment of diseases associated with orthopedic injuries, as well as to provide references for clinics.
Collapse
Affiliation(s)
- Jie Fang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China.,Graduate School of The North China University of Science and Technology, Hebei, P.R. China.,Department of Hand and Foot Surgery, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Xin Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China
| | - Wen Jiang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China
| | - Yaqiong Zhu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China
| | - Yongqiang Hu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China
| | - Yanxu Zhao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China
| | - Xueli Song
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China
| | - Jinjuan Zhao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China
| | - Wenlong Zhang
- Department of Hand and Foot Surgery, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China.,Co-innovation Center of Neuroregeneration Nantong University, Nantong, Jiangsu Province, P.R. China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, Beijing, P.R. China.,Co-innovation Center of Neuroregeneration Nantong University, Nantong, Jiangsu Province, P.R. China
| |
Collapse
|
37
|
Ehlers CB, Webb AR, McCormick BP, Wyand TJ, Sarna N, Povey K, Marano G, Schainker L. Standardized Platelet Rich Plasma Injections for Osteoarthritis of the Knee. Cureus 2020; 12:e10900. [PMID: 33062459 PMCID: PMC7550245 DOI: 10.7759/cureus.10900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective Platelet-rich plasma (PRP) and adipose-derived stem cells (ADSC) injections are non-surgical treatments for knee osteoarthritis (OA). The purpose of this study is to assess the effectiveness of serial PRP with or without ADSC injections in the treatment of refractory OA of the knee. Design Patients who failed to achieve pain relief with conventional non-surgical treatments, with Kellgren-Lawrence grade 3 or 4 knee OA, were recruited from a private outpatient clinic. Over 67 patients were elected to receive serial PRP injections and 22 patients were elected to receive an ADSC+PRP injection. These patients completed Western Ontario and McMaster Universities Arthritis Index (WOMAC) surveys prior to each treatment and at follow-up appointments. These surveys were retrospectively reviewed to assess changes in functional status and pain over time. Results Twenty-nine patients from the PRP group and eight patients in the ADSC+PRP group had adequate follow-up for inclusion in the analysis. The PRP group had an improvement in WOMAC scores by 34.30%, 60.2%, and 58.5% for patients reporting at 1-3, 4-6, and >6 months of follow-up. The ADSC+PRP group experienced an improvement of 51% at an average of 4.66 months of follow-up. Conclusions Serial PRP injections and a single ADSC+PRP injection yield improved and sustained functional outcome scores for patients with severe, refractory OA of the knee. Future studies should consider consistent orthobiologic preparation protocols to ensure reproducibility.
Collapse
Affiliation(s)
- Cooper B Ehlers
- Orthopaedics, Georgetown University School of Medicine, Washington DC, USA
| | - Alex R Webb
- Orthopaedics, Emory University School of Medicine, Atlanta, USA
| | | | - Trevor J Wyand
- Orthopaedic Surgery, Georgetown University School of Medicine, Washington DC, USA
| | - Neil Sarna
- Orthopaedics, Georgetown University School of Medicine, Washington DC, USA
| | - Kathryn Povey
- Emergency Medicine, Georgetown University School of Medicine, Washington DC, USA
| | - Geoffrey Marano
- Orthopaedics, MedStar Georgetown University Hospital, Washington DC, USA
| | - Lawrence Schainker
- Rheumatology, MedStar Georgetown University Hospital, Washington DC, USA
| |
Collapse
|
38
|
Rikkers M, Levato R, Malda J, Vonk LA. Importance of Timing of Platelet Lysate-Supplementation in Expanding or Redifferentiating Human Chondrocytes for Chondrogenesis. Front Bioeng Biotechnol 2020; 8:804. [PMID: 32733874 PMCID: PMC7360809 DOI: 10.3389/fbioe.2020.00804] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/22/2020] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) in articular joints is a prevalent disease. With increasing life expectancy, the need for therapies other than knee replacement arises. The intrinsic repair capacity of cartilage is limited, therefore alternative strategies for cartilage regeneration are being explored. The purpose of this study is first to investigate the potential of platelet lysate (PL) as a xeno-free alternative in expansion of human OA chondrocytes for cell therapy, and second to assess the effects of PL on redifferentiation of expanded chondrocytes in 3D pellet cultures. Chondrocytes were isolated from human OA cartilage and subjected to PL in monolayer culture. Cell proliferation, morphology, and expression of chondrogenic genes were assessed. Next, PL-expanded chondrocytes were cultured in 3D cell pellets and cartilage matrix production was assessed after 28 days. In addition, the supplementation of PL to redifferentiation medium for the culture of expanded chondrocytes in 3D pellets was evaluated. Glycosaminoglycan (GAG) and collagen production were evaluated by quantitative biochemical analyses, as well as by (immuno)histochemistry. A dose-dependent effect of PL on chondrocyte proliferation was found, but expression of chondrogenic markers was decreased when compared to FBS-expanded cells. After 28 days of subsequent 3D pellet culture, GAG production was significantly higher in pellets consisting of chondrocytes expanded with PL compared to controls. However, when used to supplement redifferentiation medium for chondrocyte pellets, PL significantly decreased the production of GAGs and collagen. In conclusion, chondrocyte proliferation is stimulated by PL and cartilage production in subsequent 3D culture is maintained. Furthermore, the presences of PL during redifferentiation of 3D chondrocyte strongly inhibits GAG and collagen content. The data presented in the current study indicate that while the use of PL for expansion in cartilage cell therapies is possibly beneficial, intra-articular injection of the product in the treatment of OA might be questioned.
Collapse
Affiliation(s)
- Margot Rikkers
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Luciënne A Vonk
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
39
|
Sha'ban M, Ahmad Radzi MA. Scaffolds for Cartilage Regeneration: To Use or Not to Use? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1249:97-114. [PMID: 32602093 DOI: 10.1007/978-981-15-3258-0_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Joint cartilage has been a significant focus on the field of tissue engineering and regenerative medicine (TERM) since its inception in the 1980s. Represented by only one cell type, cartilage has been a simple tissue that is thought to be straightforward to deal with. After three decades, engineering cartilage has proven to be anything but easy. With the demographic shift in the distribution of world population towards ageing, it is expected that there is a growing need for more effective options for joint restoration and repair. Despite the increasing understanding of the factors governing cartilage development, there is still a lot to do to bridge the gap from bench to bedside. Dedicated methods to regenerate reliable articular cartilage that would be equivalent to the original tissue are still lacking. The use of cells, scaffolds and signalling factors has always been central to the TERM. However, without denying the importance of cells and signalling factors, the question posed in this chapter is whether the answer would come from the methods to use or not to use scaffold for cartilage TERM. This paper presents some efforts in TERM area and proposes a solution that will transpire from the ongoing attempts to understand certain aspects of cartilage development, degeneration and regeneration. While an ideal formulation for cartilage regeneration has yet to be resolved, it is felt that scaffold is still needed for cartilage TERM for years to come.
Collapse
Affiliation(s)
- Munirah Sha'ban
- Department of Physical Rehabilitation Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia.
| | - Muhammad Aa'zamuddin Ahmad Radzi
- Department of Biomedical Science, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| |
Collapse
|
40
|
Wang M, Chen G, Li G, Wang B, Lei C. Creating Cartilage in Tissue-Engineered Chamber Using Platelet-Rich Plasma Without Cell Culture. Tissue Eng Part C Methods 2020; 26:375-383. [PMID: 32539669 DOI: 10.1089/ten.tec.2020.0049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Clinically available cartilage, such as large-volume tissue-engineered cartilage, is urgently required for various clinical applications. Tissue engineering chamber (TEC) models are a promising organ-level strategy for efficient enlargement of cells or tissues within the chamber. The conventional TEC technology is not suitable for cartilage culture, because it lacks the necessary chondrogenic growth factor, which is present in platelet-rich plasma (PRP). In this study, we added autogenous auricular cartilage fragments mixed with PRP in a TEC to obtain a large amount of engineered cartilage. Experiment: To prove the efficacy of this method, 48 New Zealand white rabbits were randomly divided into 4 groups: PRP, vascularized (Ves), PRP, PRP+Ves, and control. Auricular cartilage was harvested from the rabbits, cut into fragments (2 mm), and then injected into TECs. Cartilage constructs were harvested at week 8, and construct volumes were measured. Histological morphology, immunochemical staining, and mechanical strength were evaluated. Results: At week 8, PRP+Ves constructs developed a white, cartilage-like appearance. The volume of cartilage increased by 600% the original volume from 0.30 to 1.8 ± 0.1789 mL. Histological staining showed proliferation of edge chondrocytes in the embedded cartilage in the PRP and PRP+Ves groups. Furthermore, the cartilage constructs in the PRP+Ves group show mechanical characteristics similar to those of normal cartilage. Conclusions: Auricular cartilage fragments mixed with PRP and vascularization of the TEC showed a significantly increased cartilage tissue volume after 8 weeks of incubation in rabbits. Impact Statement Repair of defects of ear cartilage tissue has always been a huge challenge to plastic surgeons. In this article, a new method is presented to produce within 8 weeks auricular cartilage in a tissue engineering chamber without cell culture. Having such a method is a valuable step toward creating a large volume of functional cartilage tissue, which may lead to successful construction of normal auricular structure with minimal donor-site morbidity.
Collapse
Affiliation(s)
- Meishui Wang
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - Guojie Chen
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China.,Department of Burn and Plastic Surgery, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Guanmin Li
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - Biao Wang
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - Chen Lei
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| |
Collapse
|
41
|
Luo C, Xie R, Zhang J, Liu Y, Li Z, Zhang Y, Zhang X, Yuan T, Chen Y, Fan W. Low-Temperature Three-Dimensional Printing of Tissue Cartilage Engineered with Gelatin Methacrylamide. Tissue Eng Part C Methods 2020; 26:306-316. [PMID: 32349648 DOI: 10.1089/ten.tec.2020.0053] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Chunyang Luo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Xie
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiyong Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zuxi Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Yuan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yinan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weimin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Carluccio S, Martinelli D, Palamà MEF, Pereira RC, Benelli R, Guijarro A, Cancedda R, Gentili C. Progenitor Cells Activated by Platelet Lysate in Human Articular Cartilage as a Tool for Future Cartilage Engineering and Reparative Strategies. Cells 2020; 9:E1052. [PMID: 32340136 PMCID: PMC7226425 DOI: 10.3390/cells9041052] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 12/26/2022] Open
Abstract
Regenerative strategies for human articular cartilage are still challenging despite the presence of resident progenitor cell population. Today, many efforts in the field of regenerative medicine focus on the use of platelet derivatives due to their ability to reactivate endogenous mechanisms supporting tissue repair. While their use in orthopedics continues, mechanisms of action and efficacy need further characterization. We describe that the platelet lysate (PL) is able to activate chondro-progenitor cells in a terminally differentiated cartilage tissue. Primary cultures of human articular chondrocytes (ACs) and cartilage explants were set up from donor hip joint biopsies and were treated in vitro with PL. PL recruited a chondro-progenitors (CPCs)-enriched population from ex vivo cartilage culture, that showed high proliferation rate, clonogenicity and nestin expression. CPCs were positive for in vitro tri-lineage differentiation and formed hyaline cartilage-like tissue in vivo without hypertrophic fate. Moreover, the secretory profile of CPCs was analyzed, together with their migratory capabilities. Some CPC-features were also induced in PL-treated ACs compared to fetal bovine serum (FBS)-control ACs. PL treatment of human articular cartilage activates a stem cell niche responsive to injury. These facts can improve the PL therapeutic efficacy in cartilage applications.
Collapse
Affiliation(s)
- Simonetta Carluccio
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, via Leon Battista Alberti 2, 16132 Genova, Italy; (S.C.); (D.M.); (M.E.F.P.); (R.C.P.); (A.G.)
| | - Daniela Martinelli
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, via Leon Battista Alberti 2, 16132 Genova, Italy; (S.C.); (D.M.); (M.E.F.P.); (R.C.P.); (A.G.)
| | - Maria Elisabetta Federica Palamà
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, via Leon Battista Alberti 2, 16132 Genova, Italy; (S.C.); (D.M.); (M.E.F.P.); (R.C.P.); (A.G.)
| | - Rui Cruz Pereira
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, via Leon Battista Alberti 2, 16132 Genova, Italy; (S.C.); (D.M.); (M.E.F.P.); (R.C.P.); (A.G.)
- Neurobiology of miRNA, Fondazione Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Roberto Benelli
- UOSD Oncologia Molecolare e Angiogenesi, IRCCS Ospedale Policlinico San Martino, largo Rosanna Benzi 10, 16132 Genova, Italy;
| | - Ana Guijarro
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, via Leon Battista Alberti 2, 16132 Genova, Italy; (S.C.); (D.M.); (M.E.F.P.); (R.C.P.); (A.G.)
| | - Ranieri Cancedda
- Endolife S.r.l., Piazza della Vittoria 15/23, 16121 Genova, Italy;
| | - Chiara Gentili
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, via Leon Battista Alberti 2, 16132 Genova, Italy; (S.C.); (D.M.); (M.E.F.P.); (R.C.P.); (A.G.)
- Center for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV 9, 16132 Genova, Italy
| |
Collapse
|
43
|
Biological potential alterations of migratory chondrogenic progenitor cells during knee osteoarthritic progression. Arthritis Res Ther 2020; 22:62. [PMID: 32216831 PMCID: PMC7099802 DOI: 10.1186/s13075-020-2144-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/05/2020] [Indexed: 12/22/2022] Open
Abstract
Background Although increasing studies have demonstrated that chondrogenic progenitor cells (CPCs) remain present in human osteoarthritic cartilage, the biological alterations of the CPCs from the less diseased lateral tibial condyle and the more diseased medial condyle of same patient remain to be investigated. Methods CPCs were isolated from paired grade 1–2 and grade 3–4 osteoarthritic cartilage by virtue of cell migratory capacities. The cell morphology, immunophenotype, self-renewal, multi-differentiation, and cell migration of these CPCs were evaluated. Additionally, the distributions of CD105+/CD271+ cells in OA osteochondral specimen were determined. Furthermore, a high-throughput mRNA sequencing was performed. Results Migratory CPCs (mCPCs) robustly outgrew from mildly collagenases-digested osteoarthritic cartilages. The mCPCs from grade 3–4 cartilages (mCPCs, grades 3–4) harbored morphological characteristics, cell proliferation, and colony formation capacity that were similar to those of the mCPCs from the grade 1–2 OA cartilages (mCPCs, grades 1–2). However, the mCPCs (grades 3–4) highly expressed CD271. In addition, the mCPCs (grades 3–4) showed enhanced osteo-adipogenic activities and decreased chondrogenic capacity. Furthermore, the mCPCs (grades 3–4) exhibited stronger cell migration in response to osteoarthritis synovial fluids. More CD105+/CD271+ cells resided in grade 3–4 articular cartilages. Moreover, the results of mRNA sequencing showed that mCPCs (grades 3–4) expressed higher migratory molecules. Conclusions Our data suggest that more mCPCs (grades 3–4) migrate to injured articular cartilages but with enhanced osteo-adipogenic and decreased chondrogenic capacity, which might explain the pathological changes of mCPCs during the progression of OA from early to late stages. Thus, these dysfunctional mCPCs might be optional cell targets for OA therapies.
Collapse
|
44
|
Li X, Ding L, Wang Y, Li Z, Wang Q, Zhao Z, Zhao S, Wang H, Wu C, Mao N, Zhu H. Skeletal stem cell-mediated suppression on inflammatory osteoclastogenesis occurs via concerted action of cell adhesion molecules and osteoprotegerin. Stem Cells Transl Med 2019; 9:261-272. [PMID: 31774632 PMCID: PMC6988769 DOI: 10.1002/sctm.19-0300] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/04/2019] [Indexed: 01/01/2023] Open
Abstract
In the current study, we investigated how skeletal stem cells (SSCs) modulate inflammatory osteoclast (OC) formation and bone resorption. Notably, we found that intercellular adhesion molecule‐1 (ICAM‐1), vascular cell adhesion molecule‐1 (VCAM‐1), and osteoprotegerin (OPG) play a synergistic role in SSC‐mediated suppression of inflammatory osteoclastogenesis. The effect of SSCs on inflammatory osteoclastogenesis was investigated using a lipopolysaccharide‐induced mouse osteolysis model in vivo and human osteoarthritis synovial fluid (OASF) in vitro. OC formation was determined by tartrate‐resistant acid phosphatase staining. Bone resorption was evaluated by microcomputerized tomography, serum C‐terminal telopeptide assay, and pit formation assay. The expression of ICAM‐1, VCAM‐1, and OPG in SSCs and their contribution to the suppression of osteoclastogenesis were determined by flow cytometry or enzyme linked immunosorbent assay. Gene modification, neutralization antibodies, and tumor necrosis factor‐α knockout mice were used to further explore the mechanism. The results demonstrated that SSCs remarkably inhibited inflammatory osteoclastogenesis in vivo and in vitro. Mechanistically, inflammatory OASF stimulated ICAM‐1 and VCAM‐1 expression as well as OPG secretion by SSCs. In addition, ICAM‐1 and VCAM‐1 recruited CD11b+ OC progenitors to proximity with SSCs, which strengthened the inhibitory effects of SSC‐derived OPG on osteoclastogenesis. Furthermore, it was revealed that tumor necrosis factor α is closely involved in the suppressive effects. In summary, SSCs express a higher level of ICAM‐1 and VCAM‐1 and produce more OPG in inflammatory microenvironments, which are sufficient to inhibit osteoclastogenesis in a “capture and educate” manner. These results may represent a synergistic mechanism to prevent bone erosion during joint inflammation by SSCs.
Collapse
Affiliation(s)
- Xin Li
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
- Beijing Institute of Basic Medical SciencesBeijingPeople's Republic of China
- Air Force Medical Center, PLABeijingPeople's Republic of China
- Jizhong Energy Xingtai MIG General HospitalXingtaiPeople's Republic of China
| | - Li Ding
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
- Air Force Medical Center, PLABeijingPeople's Republic of China
| | - Yu‐Xing Wang
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
- People's Liberation Army General HospitalBeijingPeople's Republic of China
| | - Zhong‐Li Li
- People's Liberation Army General HospitalBeijingPeople's Republic of China
| | - Qian Wang
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
- People's Liberation Army General HospitalBeijingPeople's Republic of China
| | - Zhi‐Dong Zhao
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
- People's Liberation Army General HospitalBeijingPeople's Republic of China
| | - Sen Zhao
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
- People's Liberation Army General HospitalBeijingPeople's Republic of China
| | - Hua Wang
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
| | - Chu‐Tse Wu
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
| | - Ning Mao
- Beijing Institute of Basic Medical SciencesBeijingPeople's Republic of China
| | - Heng Zhu
- Beijing Institute of Radiation MedicineBeijingPeople's Republic of China
- Beijing Institute of Basic Medical SciencesBeijingPeople's Republic of China
| |
Collapse
|