1
|
Kumavat R, Kumar V, Biswas S. Differential Expression of Fibrinogen Alpha and Its Potential Involvement in Osteoarthritis Pathogenesis. Mol Biotechnol 2025; 67:104-114. [PMID: 38182865 DOI: 10.1007/s12033-023-00983-w] [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: 08/11/2023] [Accepted: 11/05/2023] [Indexed: 01/07/2024]
Abstract
The deterioration of cartilage tissue and other joint components composed of synovial tissue is a defining characteristic of osteoarthritis (OA) disease. Because of the lack of understanding of the underlying cause and important molecular pathways, there are currently no effective diagnostic or treatment methods for OA. The purpose of the study is to find a specific protein biomarker with high sensitivity and specificity in order to understand the pathophysiology of the disease and the underlying molecular pathways. We examined plasma samples of matched age and sex from OA patients (n = 150) and healthy controls (HC) (n = 70) to find proteins that were differentially expressed and validated by western blotting, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and immunofluorescence. The results of western blotting demonstrated that the expression level of the fibrinogen alpha (FGA) protein was higher in plasma samples of osteoarthritis (OAPL) (p = 0.0343), and the ROC (receiver operating characteristic curve) curve supported the high sensitivity (95.22%) and specificity (74%) of FGA in OA plasma compared to healthy controls. FGA protein was detected to be deposited in the synovial tissue of OA patients (p = 0.0073). By activating the Toll-like receptor (TLR-4) receptor pathway in PBMCs (p = 0.04) and synovial tissue, FGA protein may be involved in the molecular mechanism of OA pathogenesis. Our findings collectively suggested that FGA, which is significantly expressed in OA plasma, synovial tissue, and PBMCs and is connected to the disease's advancement through the TLR-4 receptor, may serve as a diagnostic or disease-evolving tool for OA.
Collapse
Affiliation(s)
- Rajkamal Kumavat
- Council of Scientific &Industrial Research (CSIR) - Institute of Genomics & Integrative Biology, Mall Road, Delhi University Campus, 110007, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vijay Kumar
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sagarika Biswas
- Council of Scientific &Industrial Research (CSIR) - Institute of Genomics & Integrative Biology, Mall Road, Delhi University Campus, 110007, Delhi, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Department of Genomics & Molecular Medicine, Institute of Genomics and Integrative Biology, New Delhi, 110007, India.
| |
Collapse
|
2
|
Scalzone A, Sanjurjo-Rodríguez C, Berlinguer-Palmini R, Dickinson AM, Jones E, Wang XN, Crossland RE. Functional and Molecular Analysis of Human Osteoarthritic Chondrocytes Treated with Bone Marrow-Derived MSC-EVs. Bioengineering (Basel) 2024; 11:388. [PMID: 38671809 PMCID: PMC11047960 DOI: 10.3390/bioengineering11040388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease, causing impaired mobility. There are currently no effective therapies other than palliative treatment. Mesenchymal stromal cells (MSCs) and their secreted extracellular vesicles (MSC-EVs) have shown promise in attenuating OA progression, promoting chondral regeneration, and modulating joint inflammation. However, the precise molecular mechanism of action driving their beneficial effects has not been fully elucidated. In this study, we analyzed MSC-EV-treated human OA chondrocytes (OACs) to assess viability, proliferation, migration, cytokine and catabolic protein expression, and microRNA and mRNA profiles. We observed that MSC-EV-treated OACs displayed increased metabolic activity, proliferation, and migration compared to the controls. They produced decreased proinflammatory (Il-8 and IFN-γ) and increased anti-inflammatory (IL-13) cytokines, and lower levels of MMP13 protein coupled with reduced expression of MMP13 mRNA, as well as negative microRNA regulators of chondrogenesis (miR-145-5p and miR-21-5p). In 3D models, MSC-EV-treated OACs exhibited enhanced chondrogenesis-promoting features (elevated sGAG, ACAN, and aggrecan). MSC-EV treatment also reversed the pathological impact of IL-1β on chondrogenic gene expression and extracellular matrix component (ECM) production. Finally, MSC-EV-treated OACs demonstrated the enhanced expression of genes associated with cartilage function, collagen biosynthesis, and ECM organization and exhibited a signature of 24 differentially expressed microRNAs, associated with chondrogenesis-associated pathways and ECM interactions. In conclusion, our data provide new insights on the potential mechanism of action of MSC-EVs as a treatment option for early-stage OA, including transcriptomic analysis of MSC-EV-treated OA, which may pave the way for more targeted novel therapeutics.
Collapse
Affiliation(s)
- Annachiara Scalzone
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Centre for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, 80125 Napoli, Italy
| | - Clara Sanjurjo-Rodríguez
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS7 4SA, UK
| | | | - Anne M. Dickinson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS7 4SA, UK
| | - Xiao-Nong Wang
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Rachel E. Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
3
|
Jeyaraman M, Nallakumarasamy A, Jeyaraman N, Ramasubramanian S. Tissue engineering in chondral defect. COMPUTATIONAL BIOLOGY FOR STEM CELL RESEARCH 2024:361-378. [DOI: 10.1016/b978-0-443-13222-3.00033-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Ahn SY, Bagheri Varzaneh M, Zhao Y, Rozynek J, Ravindran S, Banks J, Chaudhry M, Reed DA. NG2/CSPG4 attenuates motility in mandibular fibrochondrocytes under serum starvation conditions. Front Cell Dev Biol 2023; 11:1240920. [PMID: 38020894 PMCID: PMC10662293 DOI: 10.3389/fcell.2023.1240920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The migration of mandibular fibrochondrocytes is important for the development of the mandible, the homeostasis of the mandibular cartilage, and for the capacity of the tissue to respond to injury. Mandibular fibrochondrocytes have to overcome formidable obstacles during migration including a dense and heterogeneous three-dimensional matrix. Guiding the direction of cell migration and commitment to a migratory phenotype in this microenvironment necessitates a multivalent response to chemotactic and extracellular matrix-mediated stimuli. One of the key matrix components in the cartilage of the temporomandibular joint is type VI collagen. Neuron/glial antigen 2 (NG2/CSPG4) is a transmembrane proteoglycan that binds with collagen VI and has been implicated in a wide range of cell behaviors including cell migration, motility, adhesion, and proliferation. While NG2/CSPG4 has been shown to be a key regulator of mandibular cartilage homeostasis, its role in the migration of mandibular fibrochondrocytes during normal and cell stress conditions has yet to be resolved. Here, we address this gap in knowledge by characterizing NG2/CSPG4-dependent migration in mandibular fibrochondrocytes using primary mandibular fibrochondrocytes isolated from control and full length NG2/CSPG4 knockout mice, in primary mandibular fibrochondrocytes isolated from NG2|DsRed reporter mice and in an immortalized mandibular fibrochondrocyte cell line with a mutated NG2/CSPG4 ectodomain. All three cells demonstrate similar results, with loss of the full length or truncated NG2/CSPG4 increasing the rate of cell migration in serum starvation/cell stress conditions. These findings clearly implicate NG2/CSPG4 as a key molecule in the regulation of cell migration in mandibular fibrochondrocytes in normal and cell stress conditions, underscoring the role of NG2/CSPG4 as a mechanosensitive signaling hub in the mandibular cartilage.
Collapse
Affiliation(s)
- Shin Young Ahn
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Mina Bagheri Varzaneh
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Yan Zhao
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Jacob Rozynek
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Jonathan Banks
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - Minahil Chaudhry
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| | - David A. Reed
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States
| |
Collapse
|
5
|
Kang L, Yang AL, Lai CH, Chen TJ, Lin SY, Wang YH, Wang CZ, Conway EM, Wu HL, Ho ML, Chang JK, Chen CH, Cheng TL. Chondrocyte Thrombomodulin Protects against Osteoarthritis. Int J Mol Sci 2023; 24:ijms24119522. [PMID: 37298473 DOI: 10.3390/ijms24119522] [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: 05/04/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent form of arthritis that affects over 32.5 million adults worldwide, causing significant cartilage damage and disability. Unfortunately, there are currently no effective treatments for OA, highlighting the need for novel therapeutic approaches. Thrombomodulin (TM), a glycoprotein expressed by chondrocytes and other cell types, has an unknown role in OA. Here, we investigated the function of TM in chondrocytes and OA using various methods, including recombinant TM (rTM), transgenic mice lacking the TM lectin-like domain (TMLeD/LeD), and a microRNA (miRNA) antagomir that increased TM expression. Results showed that chondrocyte-expressed TM and soluble TM [sTM, like recombinant TM domain 1 to 3 (rTMD123)] enhanced cell growth and migration, blocked interleukin-1β (IL-1β)-mediated signaling and protected against knee function and bone integrity loss in an anterior cruciate ligament transection (ACLT)-induced mouse model of OA. Conversely, TMLeD/LeD mice exhibited accelerated knee function loss, while treatment with rTMD123 protected against cartilage loss even one-week post-surgery. The administration of an miRNA antagomir (miR-up-TM) also increased TM expression and protected against cartilage damage in the OA model. These findings suggested that chondrocyte TM plays a crucial role in counteracting OA, and miR-up-TM may represent a promising therapeutic approach to protect against cartilage-related disorders.
Collapse
Grants
- 110-2314-B-037-022- Ministry of Science and Technology, Executive Yuan, Taiwan
- 111-2314-B-037-055- Ministry of Science and Technology, Executive Yuan, Taiwan
- 110-2314-B-006 -037 -MY3 Ministry of Science and Technology, Executive Yuan, Taiwan
- 110-2314-B-037 -029 -MY3 Ministry of Science and Technology, Executive Yuan, Taiwan
- 111-2314-B-037-106 Ministry of Science and Technology, Executive Yuan, Taiwan
- KMTTH- 111-R002 Kaohsiung Municipal Ta-Tung Hospital
- KMTTH-DK(A)112001 Kaohsiung Municipal Ta-Tung Hospital
- KMU-TC112A02 Kaohsiung Medical University
- KMUH-DK(A)110003 Kaohsiung Medical University
- KMU-DK(B) 110002 Kaohsiung Medical University
Collapse
Affiliation(s)
- Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Ai-Lun Yang
- Institute of Sports Sciences, University of Taipei, Taipei 11153, Taiwan
| | - Chao-Han Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tsan-Ju Chen
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sung-Yen Lin
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yan-Hsiung Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chau-Zen Wang
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Edward M Conway
- Centre for Blood Research, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
| | - Mei-Ling Ho
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 800, Taiwan
| | - Je-Ken Chang
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 800, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Chung-Hwan Chen
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
6
|
Deng Z, Luo F, Lin Y, Luo J, Ke D, Song C, Xu J. Research trends of mesenchymal stem cells application in orthopedics: A bibliometric analysis of the past 2 decades. Front Public Health 2022; 10:1021818. [PMID: 36225768 PMCID: PMC9548591 DOI: 10.3389/fpubh.2022.1021818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/07/2022] [Indexed: 01/28/2023] Open
Abstract
Background Bibliometric analysis and visualization tools were used to determine the development trend of mesenchymal stem cells (MSCs) in orthopedics in the past 20 years, so as to guide researchers to explore new directions and hotspots in the field in the future. Methods In the Web of Science Core Collection, all articles about the application of MSCs in orthopedics from 2002 to 2021 were searched. The qualitative and quantitative analysis was performed based on Web of Science and CiteSpace software. Results A total of 2,207 articles were retrieved. After excluding non-article articles such as review and letter and non-English language articles, 1,489 articles were finally included. Over the past 2 decades, the number of publications on the application of MSCs in orthopedic diseases increased. Among them, the United States, China, Japan and the United Kingdom have made significant contributions in this field. The most productive institution was Shanghai Jiao Tong University. Journal of Orthopedic Research published the largest number of publications. The journal with the highest citation frequency was Experimental Hematology. The authors with the highest output and the highest citation frequency on average were Rochy S. Tuan and Scott A. Rodeo, respectively. "Mesenchymal stem cell", "in vitro" and "Differentiation" were the top three keywords that appeared. From the keyword analysis, the current research trend indicates that the primary research hotspots of MSCs in orthopedics are the source of MSCs, in vitro experiments and the differentiation of MSCs into bone and cartilage. The frontiers of this field are the combination of MSCs and platelet-rich plasma (PRP), the treatment of knee diseases such as osteoarthritis, osteogenic differentiation, and the application of biological scaffolds combined with MSCs. Conclusion Over the past 2 decades, the application of MSCs in orthopedic diseases has received increasing attention. Our bibliometric analysis results provide valuable information and research trends for researchers in the field to understand the basic knowledge of the field, identify current research hotspots, potential collaborators, and future research frontiers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jie Xu
- Department of Orthopedics, Fujian Clinical Research Center for Spinal Nerve and Joint Diseases, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| |
Collapse
|
7
|
Zhu Y, Fu W. Peripheral Blood-Derived Stem Cells for the Treatment of Cartilage Injuries: A Systematic Review. Front Bioeng Biotechnol 2022; 10:956614. [PMID: 35935493 PMCID: PMC9355401 DOI: 10.3389/fbioe.2022.956614] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The treatment of cartilage damage is a hot topic at present, and cell therapy is an emerging alternative therapy. Stem cells derived from peripheral blood have become the focus of current research due to the ease of obtaining materials and a wide range of sources.Methods: We used a text search strategy using the [“mesenchymal stem cells” (MeSH term) OR “MSC” OR “BMMSC” OR “PBMSC” OR” PBMNC” OR “peripheral blood stem cells”] AND (cartilage injury [MeSH term] OR “cartilage” OR “chondral lesion”). After searching the literature, through the inclusion and exclusion criteria, the last included articles were systematically reviewed.Result: We found that peripheral blood-derived stem cells have chondrogenic differentiation ability and can induce chondrogenic differentiation and repair in vivo and have statistical significance in clinical and imaging prognosis. It is an improvement of academic differences. Compared with the bone marrow, peripheral blood is easier to obtain, widely sourced, and simple to obtain. In the future, peripheral blood will be a more potential cell source for cell therapy in the treatment of cartilage damage.Conclusion: Stem cells derived from peripheral blood can repair cartilage and are an important resource for the treatment of cartilage damage in the future. The specific mechanism and way of repairing cartilage need further study.
Collapse
|
8
|
Song Y, Wu Z, Zhao P. The Function of Metformin in Aging-Related Musculoskeletal Disorders. Front Pharmacol 2022; 13:865524. [PMID: 35392559 PMCID: PMC8982084 DOI: 10.3389/fphar.2022.865524] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/21/2022] [Indexed: 12/22/2022] Open
Abstract
Metformin is a widely accepted first-line hypoglycemic agent in current clinical practice, and it has been applied to the clinic for more than 60 years. Recently, researchers have identified that metformin not only has an efficient capacity to lower glucose but also exerts anti-aging effects by regulating intracellular signaling molecules. With the accelerating aging process and mankind’s desire for a long and healthy life, studies on aging have witnessed an unprecedented boom. Osteoporosis, sarcopenia, degenerative osteoarthropathy, and frailty are age-related diseases of the musculoskeletal system. The decline in motor function is a problem that many elderly people have to face, and in serious cases, they may even fail to self-care, and their quality of life will be seriously reduced. Therefore, exploring potential treatments to effectively prevent or delay the progression of aging-related diseases is essential to promote healthy aging. In this review, we first briefly describe the origin of metformin and the aging of the movement system, and next review the evidence associated with its ability to extend lifespan. Furthermore, we discuss the mechanisms related to the modulation of aging in the musculoskeletal system by metformin, mainly its contribution to bone homeostasis, muscle aging, and joint degeneration. Finally, we analyze the protective benefits of metformin in aging-related diseases of the musculoskeletal system.
Collapse
Affiliation(s)
- Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Henson F, Lydon H, Birch M, Brooks R, McCaskie A. Using apheresis-derived cells to augment microdrilling in the treatment of chondral defects in an ovine model. J Orthop Res 2021; 39:1411-1422. [PMID: 33146412 PMCID: PMC7612025 DOI: 10.1002/jor.24889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/17/2020] [Accepted: 10/21/2020] [Indexed: 02/04/2023]
Abstract
The treatment of chondral defects using microdrilling often results in a mechanically weak fibrocartilagenous repair, rather than a more robust hyaline cartilage repair. Many different microfracture/microdrilling augmentation techniques have been described, including the use of cellular products to enhance healing. Autologous peripheral blood progenitor cells can be obtained via apheresis after administration of granulocyte colony-stimulating factor (G-CSF) and have been used successfully to augment microdrilling in clinical patients. The objective of this study was to use apheresis-derived mononuclear blood cells to augment microdrilling treatment of a cartilage defect in an ovine model to determine the effect on healing. Forty adult female sheep were used in this study and were divided into a control group (microdrilling alone) and a treatment group (microdrilling, hyaluronic acid, and apheretic product). Outcome measurements included weight-bearing on the operated limb, macroscopic scoring of the joint, histology, and immunohistochemistry. In addition, magnetic resonance imaging was used to attempt to identify SPION-labeled cells from the apheretic product in the operated limbs. The results showed a significant increase in healing as measured by the modified O'Driscoll sore in the treated group. No evidence of homing of SPION-labeled cells to the defect was found and no correlation was found between the response to G-CSF administration or concentration of CD34+ and outcome. A correlation was found between healing and the concentration of white blood cells and peripheral blood mononuclear cell numbers in the apheretic product.
Collapse
Affiliation(s)
- Frances Henson
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Helen Lydon
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Mark Birch
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Roger Brooks
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Andrew McCaskie
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Deng Z, Jin J, Wang S, Qi F, Chen X, Liu C, Li Y, Ma Y, Lyu F, Zheng Q. Narrative review of the choices of stem cell sources and hydrogels for cartilage tissue engineering. ANNALS OF TRANSLATIONAL MEDICINE 2021; 8:1598. [PMID: 33437797 PMCID: PMC7791208 DOI: 10.21037/atm-20-2342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Stem cell-based therapy is a promising treatment for cartilage defects due to the pluripotency, abundant sources and low immunogenicity of stem cells. Hydrogels are a promising class of biomaterials for cartilage engineering and are characterized by bioactivity, degradability and elasticity as well as provide water content and mechanical support. The combination of stem cells and hydrogels opens new possibilities for cartilage tissue engineering. However, the selection of suitable types of stem cells and hydrogels is difficult. Currently, various types of stem cells, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and peripheral blood mononuclear cells (PBMSCs), and various types of hydrogels, including natural polymers, chemically modified natural polymers and synthetic polymers, have been explored based on their potential for cartilage tissue engineering. These materials are used independently or in combination; however, there is no clear understanding of their merits and disadvantages with regard to their suitability for cartilage repair. In this article, we aim to review recent progress in the use of stem cell-hydrogel hybrid constructs for cartilage tissue engineering. We focus on the effects of stem cell types and hydrogel types on efficient chondrogenesis from cellular, preclinical and clinical perspectives. We compare and analyze the advantages and disadvantages of these cells and hydrogels with the hope of increasing discussion of their suitability for cartilage repair and present our perspective on their use for the improvement of physical and biological properties for cartilage tissue engineering.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiewen Jin
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fangjie Qi
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xuepan Chen
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chang Liu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanbing Li
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fengjuan Lyu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,South China University of Technology-the University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
11
|
Wongin S, Narkbunnam R, Waikakul S, Chotiyarnwong P, Aresanasuwan T, Roytrakul S, Viravaidya-Pasuwat K. Construction and Evaluation of Osteochondral-Like Tissue Using Chondrocyte Sheet and Cancellous Bone. Tissue Eng Part A 2020; 27:282-295. [PMID: 32718279 DOI: 10.1089/ten.tea.2020.0107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The manipulation of human chondrocyte sheets in target areas frequently results in their tearing because they are thin and fragile. In this study, human cancellous bones were used as a supporting material to create chondrocyte sheet-cancellous bone tissues, and their properties were evaluated. Using cell sheet technology, human chondrocytes were constructed into triple-layered chondrocyte sheets that displayed chondrogenic properties. After transferring the chondrocyte sheets onto cancellous bones, the top area of the chondrocyte sheet-cancellous bone tissues exhibited a smooth surface topography without cell sheet floating within 7 days of culture. The immunofluorescence staining of collagen type II (COL2A1) and fibronectin (FN1) was also performed and examined. Using the shotgun proteomic analysis, the proteins associated with cell adhesion, extracellular matrix (ECM) organization, cell-substrate junction assembly, and cell adhesion mediated by integrin were observed in the chondrocyte sheets, cancellous bones, and chondrocyte sheet-cancellous bone tissues. Three integrin members, including integrin β4 (ITGB4), ITGB6, and ITGB8, were found in the chondrocyte sheets. Only ITGB8 was found in the chondrocyte sheets and chondrocyte sheet-cancellous bone tissues. During 48 h, the mean velocity of the individual cell migration was low, which did not affect the structure and chondrogenic properties of the chondrocyte sheets. Staining of the filamentous actin (F-actin) cytoskeleton in the migratory cells also provided a better understanding of the dynamic communication between the cell cytoskeleton and adhesion molecules through ITGB8, which may play a key role in the attachment of the chondrocyte sheets and the synthesis of the cartilage ECM. Therefore, we suggest that cancellous bone could be used as a supporting material to construct chondrocyte sheet-cancellous bone tissues for potential treatment of osteochondral lesions. Impact Statement We proposed a method to construct an osteochondral-like tissue by placing human chondrocyte sheets onto cancellous bone. The stationary chondrocyte sheets and the low mean velocity of the individual cell migration on the cancellous bone with the expression of COL2A1 indicated that the cancellous bone served as an appropriate supporting material. Moreover, the cellular mechanism for the adhesion of the chondrocyte sheets on the cancellous bone based on ITGB8-mediated adhesion through the rearrangement of filamentous actin provided a better understanding to improve the construction of osteochondral-like tissues, and to predict the repair mechanism in osteoarthritis therapy.
Collapse
Affiliation(s)
- Sopita Wongin
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Rapeepat Narkbunnam
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Saranatra Waikakul
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pojchong Chotiyarnwong
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thanyawan Aresanasuwan
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathum Thani, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand.,Department of Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| |
Collapse
|
12
|
Szustak M, Gendaszewska-Darmach E. Extracellular Nucleotides Selectively Induce Migration of Chondrocytes and Expression of Type II Collagen. Int J Mol Sci 2020; 21:ijms21155227. [PMID: 32718031 PMCID: PMC7432683 DOI: 10.3390/ijms21155227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/31/2022] Open
Abstract
The migration of chondrocytes from healthy to injured tissues is one of the most important challenges during cartilage repair. Additionally, maintenance of the chondrogenic phenotype remains another limitation, especially during monolayer culture in vitro. Using both the differentiated and undifferentiated chondrogenic ATDC5 cell line, we showed that extracellular nucleotides are able to increase the migration rate of chondrocytes without affecting their chondrogenic phenotype. We checked the potency of natural nucleotides (ATP, ADP, UTP, and UDP) as well as their stable phosphorothioate analogs, containing a sulfur atom in the place of one nonbridging oxygen atom in a phosphate group. We also detected P2y1, P2y2, P2y4, P2y6, P2y12, P2y13, and P2y14 mRNA transcripts for nucleotide receptors, demonstrating that P2y1 and P2y13 are highly upregulated in differentiated ATDC5 cells. We showed that ADPβS, UDPβS, and ADP are the best stimulators of migration of differentiated chondrocytes. Additionally, ADP and ADPβS positively affected the expression of type II collagen, a structural component of the cartilage matrix.
Collapse
|
13
|
Chen YR, Yan X, Yuan FZ, Ye J, Xu BB, Zhou ZX, Mao ZM, Guan J, Song YF, Sun ZW, Wang XJ, Chen ZY, Wang DY, Fan BS, Yang M, Song ST, Jiang D, Yu JK. The Use of Peripheral Blood-Derived Stem Cells for Cartilage Repair and Regeneration In Vivo: A Review. Front Pharmacol 2020; 11:404. [PMID: 32308625 PMCID: PMC7145972 DOI: 10.3389/fphar.2020.00404] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Background Peripheral blood (PB) is a potential source of chondrogenic progenitor cells that can be used for cartilage repair and regeneration. However, the cell types, isolation and implantation methods, seeding dosage, ultimate therapeutic effect, and in vivo safety remain unclear. Methods PubMed, Embase, and the Web of Science databases were systematically searched for relevant reports published from January 1990 to December 2019. Original articles that used PB as a source of stem cells to repair cartilage in vivo were selected for analysis. Results A total of 18 studies were included. Eight human studies used autologous nonculture-expanded PB-derived stem cells (PBSCs) as seed cells with the blood cell separation isolation method, and 10 animal studies used autologous, allogenic or xenogeneic culture-expanded PB-derived mesenchymal stem cells (PB-MSCs), or nonculture-expanded PBSCs as seed cells. Four human and three animal studies surgically implanted cells, while the remaining studies implanted cells by single or repeated intra-articular injections. 121 of 130 patients (in 8 human clinical studies), and 230 of 278 animals (in 6 veterinary clinical studies) using PBSCs for cartilage repair achieved significant clinical improvement. All reviewed articles indicated that using PB as a source of seed cells enhances cartilage repair in vivo without serious adverse events. Conclusion Autologous nonculture-expanded PBSCs are currently the most commonly used cells among all stem cell types derived from PB. Allogeneic, autologous, and xenogeneic PB-MSCs are more widely used in animal studies and are potential seed cell types for future applications. Improving the mobilization and purification technology, and shortening the culture cycle of culture-expanded PB-MSCs will obviously promote the researchers' interest. The use of PBSCs for cartilage repair and regeneration in vivo are safe. PBSCs considerably warrant further investigations due to their superiority and safety in clinical settings and positive effects despite limited evidence in humans.
Collapse
Affiliation(s)
- You-Rong Chen
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Xin Yan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Fu-Zhen Yuan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Jing Ye
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Bing-Bing Xu
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Zhu-Xing Zhou
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Zi-Mu Mao
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Jian Guan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Yi-Fan Song
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Ze-Wen Sun
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China.,School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xin-Jie Wang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Ze-Yi Chen
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Ding-Yu Wang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Bao-Shi Fan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China.,School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Meng Yang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China.,School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Shi-Tang Song
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Dong Jiang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Jia-Kuo Yu
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
14
|
Stefanowicz-Hajduk J, Ochocka JR. Real-time cell analysis system in cytotoxicity applications: Usefulness and comparison with tetrazolium salt assays. Toxicol Rep 2020; 7:335-344. [PMID: 32090021 PMCID: PMC7025972 DOI: 10.1016/j.toxrep.2020.02.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 01/08/2023] Open
Abstract
RTCA system allows to easily monitor cell adhesion and proliferation. The real-time impedance technique is widely used in many toxicological studies. RTCA results are generally comparable with results from tetrazolium salts assays. RTCA analysis should be limited when drugs with electroactive additives are tested. Tetrazolium salts assays should be avoided when colored compounds are studied.
Real-time cell analysis (RTCA) is a technique based on impedance and microsensor electrodes. RTCA system allows label-free, real-time, and continuous monitoring of cell adhesion, morphology, and rate of cell proliferation. The system offers a wide range of applications, mainly in toxicological studies, new drug screening, and microbiology. Here, we describe the usefulness of the system in different applications and compare this technology with conventional endpoint assays based on tetrazolium salts. We present advantages and disadvantages of the system and endpoint methods and their limitations in cytotoxicity investigations.
Collapse
Affiliation(s)
- Justyna Stefanowicz-Hajduk
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, Al. Hallera 107, 80-416, Gdańsk, Poland
| | - J Renata Ochocka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, Al. Hallera 107, 80-416, Gdańsk, Poland
| |
Collapse
|
15
|
Hyaluronic Acid (HA), Platelet-Rich Plasm and Extracorporeal Shock Wave Therapy (ESWT) promote human chondrocyte regeneration in vitro and ESWT-mediated increase of CD44 expression enhances their susceptibility to HA treatment. PLoS One 2019; 14:e0218740. [PMID: 31251756 PMCID: PMC6599220 DOI: 10.1371/journal.pone.0218740] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/07/2019] [Indexed: 12/27/2022] Open
Abstract
Novel strategies have been proposed for articular cartilage damage occurring during osteoarthritis (OA) and -among these- Extracorporeal Shock Wave Therapy (ESWT), intra-articular injections of Platelet-Rich Plasma (PRP) or Hyaluronic Acid (HA) revealed encouraging results. To investigate the possible mechanisms responsible for those clinical benefits, we established primary cultures of human chondrocytes derived from cartilage explants and measured the in vitro effects of ESW, PRP and HA therapies. After molecular/morphological cell characterization, we assessed those effects on the functional activities of the chondrocyte cell cultures, at the protein and molecular levels. ESWT significantly prevented the progressive dedifferentiation that spontaneously occurs during prolonged chondrocyte culture. We then attested the efficiency of all such treatments to stimulate the expression of markers of chondrogenic potential such as SOX9 and COL2A, to increase the Ki67 proliferation index as well as to antagonize the traditional marker of chondrosenescence p16INK4a (known as Cdkn2a). Furthermore, all our samples showed an ESW- and HA-mediated enhancement of migratory and anti-inflammatory activity onto the cytokine-rich environment characterizing OA. Taken together, those results suggest a regenerative effect of such therapies on primary human chondrocytes in vitro. Moreover, we also show for the first time that ESW treatment induces the surface expression of major hyaluronan cell receptor CD44 allowing the increase of COL2A/COL1A ratio upon HA administration. Therefore, this work suggests that ESW-induced CD44 overexpression enhances the in vitro cell susceptibility of human chondrocytes to HA, presumably favouring the repair of degenerated cartilage.
Collapse
|
16
|
Shan J, Li S, Wang C, Liu L, Wang X, Zhu D, Fan Y, Xu J. Expression and biological functions of the CCL5‐CCR5 axis in oral lichen planus. Exp Dermatol 2019; 28:816-821. [PMID: 31006151 DOI: 10.1111/exd.13946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/02/2019] [Accepted: 04/14/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Jing Shan
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Shan Li
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
- Department of Stomatology Yancheng City No. 1 People's Hospital Yancheng China
| | - Chen Wang
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Lin Liu
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Xuewei Wang
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Dandan Zhu
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Yuan Fan
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| | - Juanyong Xu
- Jiangsu Key Laboratory of Oral Diseases Nanjing Medical University Nanjing China
- Department of Oral Medicine Affiliated Hospital of Stomatology, Nanjing Medical University Nanjing China
| |
Collapse
|
17
|
Rim YA, Nam Y, Park N, Lee J, Park SH, Ju JH. Repair potential of nonsurgically delivered induced pluripotent stem cell-derived chondrocytes in a rat osteochondral defect model. J Tissue Eng Regen Med 2018; 12:1843-1855. [PMID: 29770595 DOI: 10.1002/term.2705] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 04/13/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) are thought to be an alternative cell source for future regenerative medicine. hiPSCs may allow unlimited production of cell types that have low turnover rates and are difficult to obtain such as autologous chondrocytes. In this study, we generated hiPSC-derived chondrogenic pellets, and chondrocytes were isolated. To confirm the curative effects, chondrogenic pellets and isolated chondrocytes were transplanted into rat joints with osteochondral defects. Isolated hiPSC-derived chondrocytes were delivered in the defect by a single intra-articular injection. The generated hiPSC-derived chondrogenic pellets had increased chondrogenic marker expression and accumulated extracellular matrix proteins. Chondrocytes were successfully isolated from the pellets. Alcian blue staining and collagen type II were detected in the cells. Chondrogenic marker expression was also increased in the isolated cells. Transplanted chondrogenic pellets and chondrocytes both had curative effects in the osteochondral defect rat model. Detection of human proteins in the joints proved that the cells were successfully delivered into the defect. Chondrogenic pellets or chondrocytes generated from hiPSCs have potential as regenerative medicine for cartilage recovery or regeneration. Chondrocytes isolated from hiPSC-derived chondrogenic pellets had curative effects in damaged cartilage. Injectable hiPSC-derived chondrocytes show the possibility of noninvasive delivery of regenerative medicine for cartilage recovery.
Collapse
Affiliation(s)
- Yeri Alice Rim
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoojun Nam
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Narae Park
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jennifer Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
18
|
Lydon H, Brooks R, McCaskie A, Henson F. Peripheral mononuclear blood cell apheresis in a preclinical ovine model. BMC Vet Res 2018; 14:47. [PMID: 29439735 PMCID: PMC5812194 DOI: 10.1186/s12917-018-1332-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 01/03/2018] [Indexed: 12/26/2022] Open
Abstract
Background Recent research has demonstrated that circulating peripheral blood mononuclear fractions (PBMC) containing haematopoietic stem (HSC)/progenitor cells have the potential to play a crucial role in regenerative medicine strategies. Work in our laboratory has shown that a peripheral blood mononuclear cell fraction (PBMC) enhances cartilage repair in an osteochondral defect model in sheep and has a significant effect on cells in the joint niche. In order to obtain PBMC rich blood containing HSCs for further studies, we have performed, for the first time, apheresis on adult sheep. Results Subcutaneous granulocyte-colony stimulating factor (G-CSF) was used to mobilise white blood cells and continual flow apheresis was performed on 8 sheep under general anaesthetic. There were no observable side effects, although a marked tendency for blood clotting during the procedure was noted. The administration of G-CSF for 3 days increased the white blood cell (WBC) count in the peripheral blood from to 6.7 ± 2.1 × 106/ml to 16.1 ± 5.0 × 106/ml. Following apheresis, the WBC numbers in the apheretic product increased to 38.5 ± 27.6 × 106/ml, comprised of a significant increase in neutrophils and PBMC (from 5.25 ± 1.8 × 106/ml following G-CSF stimulation to 27.5 5 ± 27.6 × 106/ml). There was a mean of 2.1% CD34 + ve cells and 95.5% CD45 + ve cells in the apheretic product. Conclusions This study describes the administration of G-CSF and subsequent apheresis in adult sheep. The technique is safe when performed as described with no observable side effects. The technique permits collection of an increased WBC fraction containing neutrophils and PBMC in adult sheep. This apheretic product contains CD34 + ve cells, representing an HSC/progenitor population for use in in vivo and in vitro experiments.
Collapse
Affiliation(s)
- Helen Lydon
- Department of Surgery, University of Cambridge, Hill's Road, Cambridge, UK
| | - Roger Brooks
- Department of Surgery, University of Cambridge, Hill's Road, Cambridge, UK
| | - Andrew McCaskie
- Department of Surgery, University of Cambridge, Hill's Road, Cambridge, UK
| | - Frances Henson
- Department of Surgery, University of Cambridge, Hill's Road, Cambridge, UK. .,Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK.
| |
Collapse
|
19
|
Hillger JM, Lieuw WL, Heitman LH, IJzerman AP. Label-free technology and patient cells: from early drug development to precision medicine. Drug Discov Today 2017; 22:1808-1815. [PMID: 28778587 DOI: 10.1016/j.drudis.2017.07.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/10/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Drug development requires physiologically more appropriate model systems and assays to increase understanding of drug action and pathological processes in individual humans. Specifically, patient-derived cells offer great opportunities as representative cellular model systems. Moreover, with novel label-free cellular assays, it is often possible to investigate complex biological processes in their native environment. Combining these two offers distinct opportunities for increasing physiological relevance. Here, we review impedance-based label-free technologies in the context of patient samples, focusing on commonly used cell types, including fibroblasts, blood components, and stem cells. Applications extend as far as tissue-on-a-chip models. Thus, applying label-free technologies to patient samples can produce highly biorelevant data and, with them, unique opportunities for drug development and precision medicine.
Collapse
Affiliation(s)
- Julia M Hillger
- Division of Medicinal Chemistry, LACDR, Leiden University, The Netherlands
| | - Wai-Ling Lieuw
- Division of Medicinal Chemistry, LACDR, Leiden University, The Netherlands
| | - Laura H Heitman
- Division of Medicinal Chemistry, LACDR, Leiden University, The Netherlands
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, LACDR, Leiden University, The Netherlands.
| |
Collapse
|
20
|
Kimpton LS, Schwab A, Ehlicke F, Waters SL, Please CP, Whiteley JP, Byrne HM. A mathematical model for cell infiltration and proliferation in a chondral defect. Math Biosci 2017; 292:46-56. [PMID: 28735871 DOI: 10.1016/j.mbs.2017.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 05/17/2017] [Accepted: 07/18/2017] [Indexed: 11/30/2022]
Abstract
We develop a mathematical model to describe the regeneration of a hydrogel inserted into an ex vivo osteochondral explant. Specifically we use partial differential equations to describe the evolution of two populations of cells that migrate from the tissue surrounding the defect, proliferate, and compete for space and resources within the hydrogel. The two cell populations are chondrocytes and cells that infiltrate from the subchondral bone. Model simulations are used to investigate how different seeding strategies and growth factor placement within the hydrogel affect the spatial distribution of both cell types. Since chondrocyte migration is extremely slow, we conclude that the hydrogel should be seeded with chondrocytes prior to culture in order to obtain zonal chondrocyte distributions typical of those associated with healthy cartilage.
Collapse
Affiliation(s)
- L S Kimpton
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - A Schwab
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Wuerzburg, Roentgenring 11, Wuerzburg 97070, Germany
| | - F Ehlicke
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Wuerzburg, Roentgenring 11, Wuerzburg 97070, Germany
| | - S L Waters
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - C P Please
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - J P Whiteley
- Department of Computer Science, University of Oxford, Wolfson Building, Parks Road, Oxford OX1 3QD, United Kingdom.
| | - H M Byrne
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, United Kingdom
| |
Collapse
|
21
|
Abstract
OBJECTIVE Our previous study showed that WNT5A, a member of the noncanonical WNT pathway, is involved in interleukin-1beta induced matrix metalloproteinase expression in temporomandibular joint (TMJ) condylar chondrocytes. The purpose of this study is to further explore the roles of WNT5A in cartilage biology of the TMJ. METHODS An early TMJ osteoarthritis-like rat model was constructed by a mechanical method (steady mouth-opening). The gene and protein levels of WNT5A during the condylar cartilage changes were measured. Effects of WNT5A on chondrocyte proliferation, hypertrophy and migration were analyzed after WNT5A gain or loss of function in vitro. A c-Jun N-terminal kinase (JNK) inhibitor SP600125 was used to evaluate the involvement of JNK pathway in these effects of WNT5A. The expression and transcription activity of cell cycle regulators c-MYC and Cyclin D1 were examined to determine the mechanism behind WNT5A regulation of chondrocyte proliferation. RESULTS WNT5A was significantly upregulated in the condylar cartilage of rats in the early TMJ osteoarthritis-like model. Activating WNT5A facilitated condylar chondrocyte proliferation, hypertrophy and migration. Conversely, inhibiting WNT5A activity in chondrocytes decreased their proliferation, hypertrophy and migration. Blockage of the JNK pathway by its inhibitor, SP600125, impaired these effects of WNT5A on chondrocytes. WNT5A regulated both the expression and transcriptional activity of c-MYC and Cyclin D1 in chondrocytes, both of which were upregulated in condylar cartilage of the rat early TMJ osteoarthritis. CONCLUSION WNT5A regulates condylar chondrocyte proliferation, hypertrophy and migration. These findings provide new insights into the role of WNT5A signaling in TMJ cartilage biology and its potential in future therapy for TMJ degenerative diseases.
Collapse
|
22
|
Vidmar J, Chingwaru C, Chingwaru W. Mammalian cell models to advance our understanding of wound healing: a review. J Surg Res 2017; 210:269-280. [DOI: 10.1016/j.jss.2016.10.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 07/12/2016] [Accepted: 10/14/2016] [Indexed: 12/30/2022]
|
23
|
Cokelaere S, Malda J, van Weeren R. Cartilage defect repair in horses: Current strategies and recent developments in regenerative medicine of the equine joint with emphasis on the surgical approach. Vet J 2016; 214:61-71. [PMID: 27387728 DOI: 10.1016/j.tvjl.2016.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/26/2016] [Accepted: 02/07/2016] [Indexed: 12/27/2022]
Abstract
Chondral and osteochondral lesions due to injury or other pathology are highly prevalent conditions in horses (and humans) and commonly result in the development of osteoarthritis and progression of joint deterioration. Regenerative medicine of articular cartilage is an emerging clinical treatment option for patients with articular cartilage injury or disease. Functional articular cartilage restoration, however, remains a major challenge, but the field is progressing rapidly and there is an increasing body of supportive clinical and scientific evidence. This review gives an overview of the established and emerging surgical techniques employed for cartilage repair in horses. Through a growing insight in surgical cartilage repair possibilities, surgeons might be more stimulated to explore novel techniques in a clinical setting.
Collapse
Affiliation(s)
- Stefan Cokelaere
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 112, 3584 CM Utrecht, NL, Netherlands.
| | - Jos Malda
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 112, 3584 CM Utrecht, NL, Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, NL, Netherlands
| | - René van Weeren
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 112, 3584 CM Utrecht, NL, Netherlands
| |
Collapse
|
24
|
Onuora S. Regenerative medicine. PBMCs stimulate chondrocyte migration and cartilage repair. Nat Rev Rheumatol 2015; 11:563. [PMID: 26303955 DOI: 10.1038/nrrheum.2015.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|