1
|
Karimi F, Nejati B, Rahimi F, Alivirdiloo V, Alipourfard I, Aghighi A, Raji-Amirhasani A, Eslami M, Babaeizad A, Ghazi F, Firouzi Amandi A, Dadashpour M. A State-of-the-Art Review on the Recent Advances of Mesenchymal Stem Cell Therapeutic Application in Systematic Lupus Erythematosus. Immunol Invest 2024; 53:160-184. [PMID: 38031988 DOI: 10.1080/08820139.2023.2289066] [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: 12/01/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with an unknown etiology that has widespread clinical and immunological manifestations. Despite the increase in knowledge about the pathogenesis process and the increase in treatment options, however, the treatments fail in half of the cases. Therefore, there is still a need for research on new therapies. Mesenchymal stem cells (MSCs) are powerful regulators of the immune system and can reduce the symptoms of systemic lupus erythematosus. This study aimed to review the mechanisms of immune system modulation by MSCs and the role of these cells in the treatment of SLE. MSCs suppress T lymphocytes through various mechanisms, including the production of transforming growth factor-beta (TGF-B), prostaglandin E2 (PGE2), nitric oxide (NO), and indolamine 2 and 3-oxygenase (IDO). In addition, MSCs inhibit the production of their autoantibodies by inhibiting the differentiation of lymphocytes. The production of autoantibodies against nuclear antigens is an important feature of SLE. On the other hand, MSCs inhibit antigen delivery by antigen-presenting cells (APCs) to T lymphocytes. Studies in animal models have shown the effectiveness of these cells in treating SLE. However, few studies have been performed on the effectiveness of this treatment in humans. It can be expected that new treatment strategies for SLE will be introduced in the future, given the promising results of MSCs application.
Collapse
Affiliation(s)
- Farshid Karimi
- Department of Optometry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Babak Nejati
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Rahimi
- Division of Clinical Laboratory, Zahra Mardani Azar Children Training Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Alivirdiloo
- Medical Doctor Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Science, Warsaw, Poland
| | - Ali Aghighi
- Department of Clinical Biochemistry, Zahedan University of Medical Science, Zahedan, Iran
| | - Alireza Raji-Amirhasani
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Babaeizad
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Farhood Ghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mehdi Dadashpour
- Department of Medical Biotechnology, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
2
|
Wang W, Li X, Cui C, Yin G, Ren W, Wang X. Autophagy of umbilical cord mesenchymal stem cells induced by rapamycin conduces to pro-angiogenic function of the conditioned medium. Biochem Biophys Rep 2023; 36:101583. [PMID: 38053620 PMCID: PMC10694647 DOI: 10.1016/j.bbrep.2023.101583] [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: 08/10/2023] [Revised: 10/26/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023] Open
Abstract
Angiogenesis is critical for wound healing and tissue repair. Umbilical cord mesenchymal stem cells (UCMSCs)-conditioned medium has certain actions to promote angiogenesis, and is expected for wound healing and tissue repair. However, recent studies showed that the pro-angiogenic efficacy of unprocessed MSCs-conditioned medium is low, and insufficient for tissue repair. Autophagy is a process for protein recycling and a contributor for cell exocrine, which may enhance pro-angiogenic efficacy of the conditioned medium by stimulating cytokine release from UCMSCs. Therefore, in this study we attempted to obtain enhanced autophagy in UCMSCs using different concentrations of rapamycin and compared pro-angiogenic functions of the conditioned media. The in vitro data showed that although 100 nM-10 μM rapamycin all could induce autophagy in UCMSCs, 100 nM was the best dose to optimize the angiogenic effect of the conditioned medium. The in vivo data also showed that pro-angiogenic effect of the optimized conditioned medium was more obvious than that of the control conditioned medium (0 nM group) in the injected matrigel plaques. Further, the expressions of VEGF, FGF-2, MMP-9, PDGF-α and PDGF-β were markedly increased in UCMSCs treated with 100 nM rapamycin. In conclusion, appropriately enhancing autophagy of UCMSC can improve pro-angiogenic efficacy of the conditioned medium, which may optimize therapeutic applications of UCMSCs-conditioned medium in wound healing and tissue repair.
Collapse
Affiliation(s)
- Wenya Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xiao Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Chaochu Cui
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Guotian Yin
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Wenjie Ren
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
3
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
4
|
Sandanamsamy L, Harun WSW, Ishak I, Romlay FRM, Kadirgama K, Ramasamy D, Idris SRA, Tsumori F. A comprehensive review on fused deposition modelling of polylactic acid. PROGRESS IN ADDITIVE MANUFACTURING 2022; 8:1-25. [PMID: 38625345 PMCID: PMC9619022 DOI: 10.1007/s40964-022-00356-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/15/2022] [Indexed: 05/13/2023]
Abstract
Fused Deposition Modelling (FDM) is one of the additive manufacturing (AM) techniques that have emerged as the most feasible and prevalent approach for generating functional parts due to its ability to produce neat and intricate parts. FDM mainly utilises one of the widely used polymers, polylactic acid, also known as polylactide (PLA). It is an aliphatic polyester material and biocompatible thermoplastic, with the best design prospects due to its eco-friendly properties; when PLA degrades, it breaks down into water and carbon dioxide, neither of which are hazardous to the environment. However, PLA has its limitations of poor mechanical properties. Therefore, a filler reinforcement may enhance the characteristics of PLA and produce higher-quality FDM-printed parts. The processing parameters also play a significant role in the final result of the printed parts. This review aims to study and discover the properties of PLA and the optimum processing parameters. This review covers PLA in FDM, encompassing its mechanical properties, processing parameters, characterisation, and applications. A comprehensive description of FDM processing parameters is outlined as it plays a vital role in determining the quality of a printed product. In addition, PLA polymer is highly desirable for various field industrial applications such as in a medical, automobile, and electronic, given its excellent thermoplastic and biodegradability properties.
Collapse
Affiliation(s)
- L. Sandanamsamy
- Department of Mechanical Engineering, College of Engineering, Universiti Malaysia Pahang, Gambang, 26300 Kuantan, Pahang Malaysia
| | - W. S. W. Harun
- Department of Mechanical Engineering, College of Engineering, Universiti Malaysia Pahang, Gambang, 26300 Kuantan, Pahang Malaysia
| | - I. Ishak
- Faculty of Manufacturing and Mechatronic Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Malaysia
| | - F. R. M. Romlay
- Faculty of Mechanical and Automotive Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Pahang Malaysia
| | - K. Kadirgama
- Faculty of Mechanical and Automotive Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Pahang Malaysia
| | - D. Ramasamy
- Department of Mechanical Engineering, College of Engineering, Universiti Malaysia Pahang, Gambang, 26300 Kuantan, Pahang Malaysia
| | - S. R. A. Idris
- Faculty of Mechanical and Automotive Engineering Technology, Universiti Malaysia Pahang, 26600 Pekan, Pahang Malaysia
| | - F. Tsumori
- Department of Aeronautics and Astronautics, Faculty of Engineering, Kyushu University, 744 Motooka Nishi-Ku, Fukuoka, 819-0395 Japan
| |
Collapse
|
5
|
Mullen M, Williams K, LaRocca T, Duke V, Hambright WS, Ravuri SK, Bahney CS, Ehrhart N, Huard J. Mechanical strain drives exosome production, function, and miRNA cargo in C2C12 muscle progenitor cells. J Orthop Res 2022; 41:1186-1197. [PMID: 36250617 DOI: 10.1002/jor.25467] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 06/19/2022] [Accepted: 10/08/2022] [Indexed: 02/04/2023]
Abstract
Mesenchymal stem cells (MSCs) have been proven to promote tissue repair. However, concerns related to their clinical application and regulatory hurdles remain. Recent data has demonstrated the proregenerative secretome of MSCs can result in similar effects in the absence of the cells themselves. Within the secretome, exosomes have emerged as a promising regenerative component. Exosomes, which are nanosized lipid vesicles secreted by cells, encapsulate micro-RNA (miRNA), RNA, and proteins that drive MSCs regenerative potential with cell specific content. As such, there is an opportunity to optimize the regenerative potential of MSCs, and thus their secreted exosome fraction, to improve clinical efficacy. Exercise is one factor that has been shown to improve muscle progenitor cell function and regenerative potential. However, the effect of exercise on MSC exosome content and function is still unclear. To address this, we used an in vitro culture system to evaluate the effects of mechanical strain, an exercise mimetic, on C2C12 (muscle progenitor cell) exosome production and proregenerative function. Our results indicate that the total exosome production is increased by mechanical strain and can be regulated with different tensile loading regimens. Furthermore, we found that exosomes from mechanically stimulated cells increase proliferation and myogenic differentiation of naïve C2C12 cells. Lastly, we show that exosomal miRNA cargo is differentially expressed following strain. Gene ontology mapping suggests positive regulation of bone morphogenetic protein signaling, regulation of actin-filament-based processes, and muscle cell apoptosis may be at least partially responsible for the proregenerative effects of exosomes from mechanically stimulated C2C12 muscle progenitor cells.
Collapse
Affiliation(s)
- Michael Mullen
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, Colorado, USA
| | - Katherine Williams
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Tom LaRocca
- Deptartment of Health and Exercise Science, Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, USA
| | - Victoria Duke
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, Colorado, USA
| | - William S Hambright
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, Colorado, USA
| | - Sudheer K Ravuri
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, Colorado, USA
| | - Chelsea S Bahney
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, Colorado, USA.,Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA.,Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital (ZSFG), University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Nicole Ehrhart
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA.,Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, USA
| | - Johnny Huard
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, Colorado, USA.,Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
6
|
Bone marrow aspirate concentrate quality is affected by age and harvest site. Knee Surg Sports Traumatol Arthrosc 2022; 31:2140-2151. [PMID: 36156111 PMCID: PMC10183435 DOI: 10.1007/s00167-022-07153-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE To compare the number and properties of bone marrow stromal cells (BMSCs) collected from bone marrow aspirate concentrate (BMAC) obtained from different harvest sites and from patients of different ages. METHODS BMAC was obtained from two groups of patients based on age (n = 10 per group): 19.0 ± 2.7 years for the younger and 56.8 ± 12.5 for the older group. In the latter, BMAC was obtained from both iliac crest and proximal tibia for a donor-matched analysis. Mononucleated cell count and CFU-F assay were performed, together with phenotype characterization of BMSCs from iliac crest and proximal tibia, the study of chondrogenic and osteogenic differentiation capacity, histological staining and spectrophotometric quantification, and the analysis of mRNAs expression. RESULTS Cells derived from iliac crest and proximal tibia showed the same phenotypic pattern at flow cytometry, as well as similar chondrogenic and osteogenic potential. However, a significantly higher number of mononuclear cells per ml was observed in younger patients (3.8 ± 1.8 × 107) compared to older patients (1.2 ± 0.8 × 107) (p < 0.0005). The latter yield, obtained from the iliac crest, was significantly higher than resulting from the BMAC harvested from the proximal tibia in the same group of patients (0.3 ± 0.2 × 107, p < 0.0005). This result was confirmed by the CFU-F analysis at day 10 (15.9 ± 19.4 vs 0.6 ± 1.0, p = 0.001) and day-20 (21.7 ± 23.0 vs 2.9 ± 4.2, p = 0.006). CONCLUSION Harvest site and age can affect the quality of BMAC. BMSCs obtained from iliac crest and proximal tibia present comparable mesenchymal markers expression as well as osteogenic and chondrogenic differentiation potential, but iliac crest BMAC presents a four times higher number of mononucleated cells with significantly higher clonogenic capacity compared to the tibia. BMAC of younger patients also had a three-time higher number of mononucleated cells. The identification of BMAC characteristics could help to optimize its preparation and to identify the most suitable indications for this orthobiologic treatment in the clinical practice.
Collapse
|
7
|
Intradiscal Therapies for Lumbar Degenerative Disk Disease. J Am Acad Orthop Surg 2022; 30:e1084-e1094. [PMID: 35984081 DOI: 10.5435/jaaos-d-21-01155] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
Discogenic low back pain is a common musculoskeletal complaint in patients presenting to orthopaedic surgeons. In addition to surgical options, there are several nonsurgical intradiscal treatments that have gained interest, ranging from biologic, nonbiologic, cell-based, and molecular therapies. However, there is limited evidence for many of these techniques, and some are still in the clinical trial stage. We describe a broad overview of these intradiscal therapies, the mechanism of action, and the evidence behind them.
Collapse
|
8
|
Autologous Stem Cells for the Treatment of Chondral Injury and Disease. OPER TECHN SPORT MED 2022. [DOI: 10.1016/j.otsm.2022.150963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
9
|
Dubay R, Urban JN, Darling EM. Single-Cell Microgels for Diagnostics and Therapeutics. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2009946. [PMID: 36329867 PMCID: PMC9629779 DOI: 10.1002/adfm.202009946] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Indexed: 05/14/2023]
Abstract
Cell encapsulation within hydrogel droplets is transforming what is feasible in multiple fields of biomedical science such as tissue engineering and regenerative medicine, in vitro modeling, and cell-based therapies. Recent advances have allowed researchers to miniaturize material encapsulation complexes down to single-cell scales, where each complex, termed a single-cell microgel, contains only one cell surrounded by a hydrogel matrix while remaining <100 μm in size. With this achievement, studies requiring single-cell resolution are now possible, similar to those done using liquid droplet encapsulation. Of particular note, applications involving long-term in vitro cultures, modular bioinks, high-throughput screenings, and formation of 3D cellular microenvironments can be tuned independently to suit the needs of individual cells and experimental goals. In this progress report, an overview of established materials and techniques used to fabricate single-cell microgels, as well as insight into potential alternatives is provided. This focused review is concluded by discussing applications that have already benefited from single-cell microgel technologies, as well as prospective applications on the cusp of achieving important new capabilities.
Collapse
Affiliation(s)
- Ryan Dubay
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
- Draper, 555 Technology Sq., Cambridge, MA 02139, USA
| | - Joseph N Urban
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
| | - Eric M Darling
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Center for Biomedical Engineering, School of Engineering, Department of Orthopaedics, Brown University, 175 Meeting St., Providence, RI 02912, USA
| |
Collapse
|
10
|
Nino-Fong R, Esparza Gonzalez BP, Rodriguez-Lecompte JC, Montelpare W, McDuffee L. Development of a biologically immortalized equine stem cell line. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2021; 85:293-301. [PMID: 34602734 PMCID: PMC8451704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/16/2021] [Indexed: 06/13/2023]
Abstract
Bone repair in horses implies invasive surgeries and increased cost. Research on musculoskeletal disorders therapy in horses includes cell-based therapy with mesenchymal stromal cells (MSCs). Mesenchymal stromal cells can be obtained from bone marrow (BMMSCs). Unfortunately, BMMSCs have limited cell replication in vitro. The objective of this study was to develop a biologically immortalized equine stem cell line derived from bone marrow, with unlimited in-vitro proliferation and the ability to differentiate into bone cells. Equine BMMSCs were transfected and immortalized with human telomerase reverse transcriptase (hTERT) gene. Cell passages from equine immortal BMMSCs were characterized by the presence of stemness CD markers and expression of multi-potent differentiation genes (OCT-4, SOX2, and NANOG). Equine immortal BMMSCs were incubated in osteogenic medium and bone cell differentiation was determined by alkaline phosphatase and von Kossa staining, and osteogenic gene expression (osteocalcin, Runx2, and osterix). Telomerase activity was determined by telomeric repeat amplification technique. Results showed that equine immortal BMMSCs were able to replicate in-vitro up to passage 50 and maintain stem cell characteristics by the presence of CD90 and expression of multi-potent genes. Equine immortal BMMSCs were able to differentiate into bone cells, which was confirmed by the positive osteogenic staining and gene expression. Equine BMMSCs were successfully immortalized and maintained characteristics of stem cells and readily differentiated into osteogenic cells. Extending the life span of equine BMMSCs by transfection of the hTERT gene will revolutionize the clinical use of MSCs by making them available to orthopedic surgeons "off the shelf."
Collapse
Affiliation(s)
- Rodolfo Nino-Fong
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, New York 11548, USA (Nino-Fong); Department of Health Management (Esparza Gonzalez, McDuffee), Department of Pathology and Microbiology (Rodriguez-Lecompte), and Department of Applied Human Sciences (Montelpare), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| | - Blanca P Esparza Gonzalez
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, New York 11548, USA (Nino-Fong); Department of Health Management (Esparza Gonzalez, McDuffee), Department of Pathology and Microbiology (Rodriguez-Lecompte), and Department of Applied Human Sciences (Montelpare), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| | - Juan Carlos Rodriguez-Lecompte
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, New York 11548, USA (Nino-Fong); Department of Health Management (Esparza Gonzalez, McDuffee), Department of Pathology and Microbiology (Rodriguez-Lecompte), and Department of Applied Human Sciences (Montelpare), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| | - William Montelpare
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, New York 11548, USA (Nino-Fong); Department of Health Management (Esparza Gonzalez, McDuffee), Department of Pathology and Microbiology (Rodriguez-Lecompte), and Department of Applied Human Sciences (Montelpare), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| | - Laurie McDuffee
- Department of Biomedical Sciences, College of Veterinary Medicine, Long Island University, 720 Northern Boulevard, Brookville, New York 11548, USA (Nino-Fong); Department of Health Management (Esparza Gonzalez, McDuffee), Department of Pathology and Microbiology (Rodriguez-Lecompte), and Department of Applied Human Sciences (Montelpare), Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3
| |
Collapse
|
11
|
Branch EA, Matuska AM, Plummer HA, Harrison RM, Anz AW. Platelet-Rich Plasma Devices Can Be Used to Isolate Stem Cells From Synovial Fluid at the Point of Care. Arthroscopy 2021; 37:893-900. [PMID: 33010328 DOI: 10.1016/j.arthro.2020.09.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 09/12/2020] [Accepted: 09/19/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE To assess whether point-of-care devices designed for collecting cellular components from blood or bone marrow could be used to isolate viable stem cells from synovial fluid. METHODS Male and female patients older than 18 years old with either an acute, anterior cruciate ligament (ACL) injury or knee osteoarthritis (OA) with a minimum estimated 20 mL of knee effusion volunteered. Ten patients with an ACL injury and 10 patients with OA were enrolled. Two milliliters of collected synovial effusion were analyzed and cultured for cellular content. The remaining fluid was combined with whole blood and processed using a buffy-coat based platelet-rich plasma (PRP) processing system. Specimens were analyzed for cell counts, colony-forming unit (CFU) assays, differentiation assays, and flow cytometry. RESULTS ACL effusion fluid contained 42.1 ± 20.7 CFU/mL and OA effusion fluid contained 65.4 ± 42.1 CFU/mL. After PRP processing, the counts in ACL-PRP were 101.6 ± 66.1 CFU/mL and 114.8 ± 73.4 CFU/mL in the OA-PRP. Cells showed tri-lineage differentiation potential when cultured under appropriate parameters. When analyzed with flow cytometry, >95% of cells produced with culturing expressed cell surface markers typically expressed by known stem cell populations, specifically CD45-, CD73+, CD29+, CD44+, CD105+, and CD90+. CONCLUSIONS Multipotent viable stem cells can be harvested from knee synovial fluid, associated with an ACL injury or OA, and concentrated with a buffy coat-based PRP-processing device. CLINICAL RELEVANCE PRP devices can be used to harvest stem cells from effusion fluids. Methods to use effusion fluid associated with an ACL injury and OA should be investigated further.
Collapse
Affiliation(s)
- Eric A Branch
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A
| | | | - Hillary A Plummer
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A
| | | | - Adam W Anz
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A..
| |
Collapse
|
12
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
13
|
18F- based Quantification of the Osteogenic Potential of hMSCs. Int J Mol Sci 2020; 21:ijms21207692. [PMID: 33080871 PMCID: PMC7589629 DOI: 10.3390/ijms21207692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
In bone tissue engineering, there is a constant need to design new methods for promoting in vitro osteogenic differentiation. Consequently, there is a strong demand for fast, effective and reliable methods to track and quantify osteogenesis in vitro. In this study, we used the radiopharmacon fluorine-18 (18F) to evaluate the amount of hydroxylapatite produced by mesenchymal stem cells (MSCs) in a monolayer cell culture in vitro. The hydroxylapatite bound tracer was evaluated using µ-positron emission tomography (µ-PET) scanning and activimeter analysis. It was therefore possible to determine the amount of synthesized mineral and thus to conclude the osteogenic potential of the cells. A Student's t-test revealed a highly significant difference regarding tracer uptake between the osteogenic group and the corresponding control group (µ-PET p = 0.043; activimeter analysis p = 0.012). This tracer uptake showed a highly significant correlation with the gold standard of quantitative Alizarin Red staining (ARS) (r2 = 0.86) as well as with the absolute calcium content detected by inductively coupled plasma mass spectrometry (r2 = 0.81). The results showed that 18F labeling is a novel method to prove and quantify hydroxyapatite content in MSC monolayer cultures. The mineral layer remains intact for further analysis. This non-destructive in vitro method can be used to rapidly investigate bone tissue engineering strategies in terms of hydroxylapatite production, and could therefore accelerate the process of implementing new strategies in clinical practice.
Collapse
|
14
|
Abstract
Regenerative medicine is gaining more and more space for the treatment of Achilles pathologic conditions. Biologics could play a role in the management of midportion Achilles tendinopathy as a step between conservative and surgical treatment or as an augmentation. Higher-level studies are needed before determining a level of treatment recommendation for biologic strategies for insertional Achilles tendinopathy. Combining imaging with patient's functional requests could be the way to reach a protocol for the use of biologics for the treatment of midportion Achilles tendinopathy and, for this perspective, the authors describe the Foot and Ankle Reconstruction Group algorithm of treatment.
Collapse
Affiliation(s)
- Cristian Indino
- IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi, 4, Milan 20161, Italy.
| | - Riccardo D'Ambrosi
- IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi, 4, Milan 20161, Italy
| | - Federico G Usuelli
- Humanitas San Pio X, via Francesco Nava, 31, 20159 Milano, Lombardia, Italy
| |
Collapse
|
15
|
Kondo S, Nakagawa Y, Mizuno M, Katagiri K, Tsuji K, Kiuchi S, Ono H, Muneta T, Koga H, Sekiya I. Transplantation of Aggregates of Autologous Synovial Mesenchymal Stem Cells for Treatment of Cartilage Defects in the Femoral Condyle and the Femoral Groove in Microminipigs. Am J Sports Med 2019; 47:2338-2347. [PMID: 31306591 DOI: 10.1177/0363546519859855] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Previous work has demonstrated that patients with cartilage defects of the knee benefit from arthroscopic transplantation of autologous synovial mesenchymal stem cells (MSCs) in terms of magnetic resonance imaging (MRI), qualitative histologic findings, and Lysholm score. However, the effectiveness was limited by the number of cells obtained, so large-sized defects (>500 mm2) were not investigated. The use of MSC aggregates may enable treatment of larger defects by increasing the number of MSCs adhering to the cartilage defect. PURPOSE To investigate whether transplantation of aggregates of autologous synovial MSCs with 2-step surgery could promote articular cartilage regeneration in microminipig osteochondral defects. STUDY DESIGN Controlled laboratory study. METHODS Synovial MSCs derived from a microminipig were examined for in vitro colony-forming and multidifferentiation abilities. An aggregate of 250,000 synovial MSCs was formed with hanging drop culture, and 16 aggregates (for each defect) were implanted on both osteochondral defects (6 × 6 × 1.5 mm) created in the medial femoral condyle and femoral groove (MSC group). The defects in the contralateral knee were left empty (control group). The knee joints were evaluated at 4 and 12 weeks by macroscopic findings and histology. MRI T1rho mapping images were acquired at 12 weeks. For cell tracking, synovial MSCs were labeled with ferucarbotran before aggregate formation and were observed with MRI at 1 week. RESULTS Synovial MSCs showed in vitro colony-forming and multidifferentiation abilities. Regenerative cartilage formation was significantly better in the MSC group than in the control group, as indicated by International Cartilage Repair Society score (macro), modified Wakitani score (histology), and T1rho mapping (biochemical MRI) in the medial condyle at 12 weeks. Implanted cells, labeled with ferucarbotran, were observed in the osteochondral defects at 1 week with MRI. No significant difference was noted in the modified Wakitani score at 4 weeks in the medial condyle and at 4 and 12 weeks in the femoral groove. CONCLUSION Transplantation of autologous synovial MSC aggregates promoted articular cartilage regeneration at the medial femoral condyle at 12 weeks in microminipigs. CLINICAL RELEVANCE Aggregates of autologous synovial MSCs could expand the indications for cartilage repair with synovial MSCs.
Collapse
Affiliation(s)
- Shimpei Kondo
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Nakagawa
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuru Mizuno
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenta Katagiri
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunikazu Tsuji
- Department of Cartilage Regeneration, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | - Takeshi Muneta
- National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
16
|
Singhvi MS, Zinjarde SS, Gokhale DV. Polylactic acid: synthesis and biomedical applications. J Appl Microbiol 2019; 127:1612-1626. [PMID: 31021482 DOI: 10.1111/jam.14290] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/29/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
Social and economic development has driven considerable scientific and engineering efforts on the discovery, development and utilization of polymers. Polylactic acid (PLA) is one of the most promising biopolymers as it can be produced from nontoxic renewable feedstock. PLA has emerged as an important polymeric material for biomedical applications on account of its properties such as biocompatibility, biodegradability, mechanical strength and process ability. Lactic acid (LA) can be obtained by fermentation of sugars derived from renewable resources such as corn and sugarcane. PLA is thus an eco-friendly nontoxic polymer with features that permit use in the human body. Although PLA has a wide spectrum of applications, there are certain limitations such as slow degradation rate, hydrophobicity and low impact toughness associated with its use. Blending PLA with other polymers offers convenient options to improve associated properties or to generate novel PLA polymers/blends for target applications. A variety of PLA blends have been explored for various biomedical applications such as drug delivery, implants, sutures and tissue engineering. PLA and their copolymers are becoming widely used in tissue engineering for function restoration of impaired tissues due to their excellent biocompatibility and mechanical properties. The relationship between PLA material properties, manufacturing processes and development of products with desirable characteristics is described in this article. LA production, PLA synthesis and their applications in the biomedical field are also discussed.
Collapse
Affiliation(s)
- M S Singhvi
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - S S Zinjarde
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - D V Gokhale
- CSIR-National Chemical Laboratory, NCIM Resource Centre, Pune, India
| |
Collapse
|
17
|
Wang G, Man Z, Xin H, Li Y, Wu C, Sun S. Enhanced adhesion and proliferation of bone marrow mesenchymal stem cells on β‑tricalcium phosphate modified by an affinity peptide. Mol Med Rep 2018; 19:375-381. [PMID: 30431109 PMCID: PMC6297790 DOI: 10.3892/mmr.2018.9655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/09/2018] [Indexed: 11/09/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are often used in orthopedic tissue engineering, and bone marrow-derived mesenchymal stem cells (BMSCs) are currently considered the gold standard. One of the most important issues in MSC-based tissue engineering therapy is the low number of MSCs in pathological tissues. Achieving efficient recruitment of MSCs to defective or damaged tissues in vivo has been a difficult hurdle. The aim of the present study was to construct a biomaterial that can effectively recruit BMSCs to facilitate the repair of pathological tissues. So functional β-tricalcium phosphate (β-TCP) was synthesized using the BMSC affinity peptide DPIYALSWSGMA (DPI) adsorbed onto β-TCP through an adsorption/freeze-drying strategy. C57BL/6 mouse-derived BMSCs were seeded onto the DPI peptide-modified β-TCP (β-TCP-DPI); in vitro experiments demonstrated that β-TCP-DPI enhanced BMSC adhesion and proliferation compared with unmodified β-TCP. Results from the present study indicated that functional β-TCP may be used as an ideal scaffold in tissue engineering and in regenerative medicine.
Collapse
Affiliation(s)
- Guozong Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hua Xin
- Department of Neurology, People's Hospital of Rizhao, Rizhao, Shandong 222000, P.R. China
| | - Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Changshun Wu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
18
|
Injectable Systems for Intra-Articular Delivery of Mesenchymal Stromal Cells for Cartilage Treatment: A Systematic Review of Preclinical and Clinical Evidence. Int J Mol Sci 2018. [PMID: 30366400 DOI: 10.3390/ijms19113322.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Stem cell-based therapy is a promising approach to treat cartilage lesions and clinical benefits have been reported in a number of studies. However, the efficacy of cell injection procedures may be impaired by cell manipulation and damage as well as by cell dissemination to non-target tissues. To overcome such issues, mesenchymal stromal cell (MSC) delivery may be performed using injectable vehicles as containment systems that further provide a favorable cell microenvironment. The aim of this systematic review was to analyze the preclinical and clinical literature on platelet-rich plasma (PRP), hyaluronic acid (HA), and hydrogels for the delivery of MSCs. The systematic literature search was performed using the PubMed and Web of science databases with the following string: "(stem cells injection) AND (platelet rich plasma OR PRP OR platelet concentrate OR biomaterials OR hyaluronic acid OR hydrogels)": 40 studies (19 preclinical and 21 clinical) met the inclusion criteria. This review revealed an increasing interest on the use of injectable agents for MSC delivery. However, while negligible adverse events and promising clinical outcomes were generally reported, the prevalence of low quality studies hinders the possibility to demonstrate the real benefits of using such injectable systems. Specific studies must be designed to clearly demonstrate the added benefits of these systems to deliver MSCs for the treatment of cartilage lesions and osteoarthritis.
Collapse
|
19
|
Roffi A, Nakamura N, Sanchez M, Cucchiarini M, Filardo G. Injectable Systems for Intra-Articular Delivery of Mesenchymal Stromal Cells for Cartilage Treatment: A Systematic Review of Preclinical and Clinical Evidence. Int J Mol Sci 2018; 19:ijms19113322. [PMID: 30366400 PMCID: PMC6274908 DOI: 10.3390/ijms19113322] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/16/2022] Open
Abstract
Stem cell-based therapy is a promising approach to treat cartilage lesions and clinical benefits have been reported in a number of studies. However, the efficacy of cell injection procedures may be impaired by cell manipulation and damage as well as by cell dissemination to non-target tissues. To overcome such issues, mesenchymal stromal cell (MSC) delivery may be performed using injectable vehicles as containment systems that further provide a favorable cell microenvironment. The aim of this systematic review was to analyze the preclinical and clinical literature on platelet-rich plasma (PRP), hyaluronic acid (HA), and hydrogels for the delivery of MSCs. The systematic literature search was performed using the PubMed and Web of science databases with the following string: "(stem cells injection) AND (platelet rich plasma OR PRP OR platelet concentrate OR biomaterials OR hyaluronic acid OR hydrogels)": 40 studies (19 preclinical and 21 clinical) met the inclusion criteria. This review revealed an increasing interest on the use of injectable agents for MSC delivery. However, while negligible adverse events and promising clinical outcomes were generally reported, the prevalence of low quality studies hinders the possibility to demonstrate the real benefits of using such injectable systems. Specific studies must be designed to clearly demonstrate the added benefits of these systems to deliver MSCs for the treatment of cartilage lesions and osteoarthritis.
Collapse
Affiliation(s)
- Alice Roffi
- Laboratory of Nano-Biotechnology-IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Norimasa Nakamura
- Institute for Medical Science in Sports, Osaka Health Science University, Osaka 590-0496, Japan.
| | - Mikel Sanchez
- Arthroscopic Surgery Unit-UCA, Hospital Vithas San Jose, 01008 Vitoria-Gasteiz, Spain.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, 66421 Homburg/Saar, Germany.
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center-IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| |
Collapse
|
20
|
Khedgikar V, Lehoczky JA. Evidence for Lgr6 as a Novel Marker of Osteoblastic Progenitors in Mice. JBMR Plus 2018; 3:e10075. [PMID: 30828690 DOI: 10.1002/jbm4.10075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/25/2018] [Accepted: 08/05/2018] [Indexed: 11/07/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells are an important source of osteoblasts critical for both bone homeostasis and repair. The ability to isolate, or specifically target, mesenchymal stem cells committed to the osteogenic lineage is necessary for orthopedic translational therapy efforts; however the precise molecular signature of these cells remains elusive. Previously, we identified a population of osteoprogenitor cells expressing the Wnt signaling agonist Lgr6, which contributes to the development and regeneration of the mouse digit tip bone. In our present study we build upon this data and investigate the expression of Lgr6 more broadly in the skeleton. We find that Lgr6, and closely related Lgr4, are expressed in mouse primary calvarial cells, bone marrow cells, and bone marrow-derived mesenchymal stem cells. In addition, our data demonstrates that Lgr4 expression is modestly increased throughout the differentiation and mineralization of mesenchymal stem cells. In contrast, we find Lgr6 expression to be strikingly increased upon osteogenic induction and subsequently decreased upon differentiation and mineralization. These findings provide evidence for Lgr6 as a novel marker of osteoprogenitor cells in bone marrow, which could prove useful for isolation of this population toward future research and clinical applications.
Collapse
Affiliation(s)
- Vikram Khedgikar
- Department of Orthopedic Surgery Brigham and Women's Hospital Harvard Medical School Boston MA USA
| | - Jessica A Lehoczky
- Department of Orthopedic Surgery Brigham and Women's Hospital Harvard Medical School Boston MA USA
| |
Collapse
|
21
|
Cotter EJ, Wang KC, Yanke AB, Chubinskaya S. Bone Marrow Aspirate Concentrate for Cartilage Defects of the Knee: From Bench to Bedside Evidence. Cartilage 2018; 9:161-170. [PMID: 29126349 PMCID: PMC5871125 DOI: 10.1177/1947603517741169] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective To critically evaluate the current basic science, translational, and clinical data regarding bone marrow aspirate concentrate (BMAC) in the setting of focal cartilage defects of the knee and describe clinical indications and future research questions surrounding the clinical utility of BMAC for treatment of these lesions. Design A literature search was performed using the PubMed and Ovid MEDLINE databases for studies in English (1980-2017) using keywords, including ["bone marrow aspirate" and "cartilage"], ["mesenchymal stem cells" and "cartilage"], and ["bone marrow aspirate" and "mesenchymal stem cells" and "orthopedics"]. A total of 1832 articles were reviewed by 2 independent authors and additional literature found through scanning references of cited articles. Results BMAC has demonstrated promising results in the clinical application for repair of chondral defects as an adjuvant procedure or as an independent management technique. A subcomponent of BMAC, bone marrow derived-mesenchymal stem cells (MSCs) possess the ability to differentiate into cells important for osteogenesis and chondrogenesis. Modulation of paracrine signaling is perhaps the most important function of BM-MSCs in this setting. In an effort to increase the cellular yield, authors have shown the ability to expand BM-MSCs in culture while maintaining phenotype. Conclusions Translational studies have demonstrated good clinical efficacy of BMAC both concomitant with cartilage restoration procedures, at defined time points after surgery, and as isolated injections. Early clinical data suggests BMAC may help stimulate a more robust hyaline cartilage repair tissue response. Numerous questions remain regarding BMAC usage, including cell source, cell expansion, optimal pathology, and injection timing and quantity.
Collapse
Affiliation(s)
- Eric J. Cotter
- Georgetown University School of Medicine, Washington, DC, USA
| | - Kevin C. Wang
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Adam B. Yanke
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
22
|
Chen YH, Chung CC, Liu YC, Lai WC, Lin ZS, Chen TM, Li LY, Hung MC. YY1 and HDAC9c transcriptionally regulate p38-mediated mesenchymal stem cell differentiation into osteoblasts. Am J Cancer Res 2018; 8:514-525. [PMID: 29637005 PMCID: PMC5883100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 02/20/2018] [Indexed: 06/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have a high self-renewal potential and can differentiate into various types of cells, including adipocytes, osteoblasts, and chondrocytes. Previously, we reported that the enhancer of zeste homolog 2 (EZH2), the catalytic component of the Polycomb-repressive complex 2, and HDAC9c mediate the osteogenesis and adipogenesis of MSCs. In the current study, we identify the role of p38 in osteogenic differentiation from a MAPK antibody array screen and investigate the mechanisms underlying its transcriptional regulation. Our data show that YY1, a ubiquitously expressed transcription factor, and HDAC9c coordinate p38 transcriptional activity to promote its expression to facilitate the osteogenic potential of MSCs. Our results show that p38 mediates osteogenic differentiation, and this has significant implications in bone-related diseases, bone tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Ya-Huey Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical UniversityTaichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University HospitalTaichung 40447, Taiwan
- Cancer Biology and Drug Discovery Ph.D. Program, China Medical UniversityTaichung 40447, Taiwan
| | - Chiao-Chen Chung
- Center for Molecular Medicine, China Medical University HospitalTaichung 40447, Taiwan
| | - Yu-Chia Liu
- Center for Molecular Medicine, China Medical University HospitalTaichung 40447, Taiwan
| | - Wei-Chen Lai
- Center for Molecular Medicine, China Medical University HospitalTaichung 40447, Taiwan
| | - Zong-Shin Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical UniversityTaichung 40402, Taiwan
| | - Tsung-Ming Chen
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and TechnologyKaohsiung 81157, Taiwan
| | - Long-Yuan Li
- Center for Molecular Medicine, China Medical University HospitalTaichung 40447, Taiwan
- Cancer Biology and Drug Discovery Ph.D. Program, China Medical UniversityTaichung 40447, Taiwan
- Department of Life Sciences, National Chung Hsing UniversityTaichung 40227, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical UniversityTaichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University HospitalTaichung 40447, Taiwan
- Cancer Biology and Drug Discovery Ph.D. Program, China Medical UniversityTaichung 40447, Taiwan
- Department of Biotechnology, Asia UniversityTaichung 41354, Taiwan
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer CenterHouston, Texas 77030, USA
| |
Collapse
|
23
|
Lebaschi A, Nakagawa Y, Wada S, Cong GT, Rodeo SA. Tissue-specific endothelial cells: a promising approach for augmentation of soft tissue repair in orthopedics. Ann N Y Acad Sci 2018; 1410:44-56. [PMID: 29265420 DOI: 10.1111/nyas.13575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/12/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
Biologics are playing an increasingly significant role in the practice of modern medicine and surgery in general and orthopedics in particular. Cell-based approaches are among the most important and widely used modalities in orthopedic biologics, with mesenchymal stem cells and other multi/pluripotent cells undergoing evaluation in numerous preclinical and clinical studies. On the other hand, fully differentiated endothelial cells (ECs) have been found to perform critical roles in homeostasis of visceral tissues through production of an adaptive panel of so-called "angiocrine factors." This newly discovered function of ECs renders them excellent candidates for novel approaches in cell-based biologics. Here, we present a review of the role of ECs and angiocrine factors in some visceral tissues, followed by an overview of current cell-based approaches and a discussion of the potential applications of ECs in soft tissue repair.
Collapse
Affiliation(s)
- Amir Lebaschi
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Yusuke Nakagawa
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Susumu Wada
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Guang-Ting Cong
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York
| | - Scott A Rodeo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York.,Sports Medicine and Shoulder Service, Hospital for Special Surgery, New York, New York
| |
Collapse
|
24
|
Sansone V, Brañes M, Romeo P. A novel bimodal approach for treating atrophic bone non-unions with extracorporeal shockwaves and autologous mesenchymal stem cell transplant. Med Hypotheses 2017; 111:4-7. [PMID: 29406993 DOI: 10.1016/j.mehy.2017.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/07/2017] [Accepted: 12/08/2017] [Indexed: 12/11/2022]
Abstract
We propose a novel approach for the treatment of atrophic bone non-unions via parallel applications of extracorporeal shock wave therapy (ESWT) and an autologous mesenchymal stem cell transplant. The hypothesis resides on the potentiality of shock waves (SWs) to act as a tool for manipulating the patient's mesenchymal stem cells (MSCs). In addition to the conventional physical stimulus achieved by delivering SWs at the site of non-union to stimulate the well-known trophic effects on bone tissue, a series of concomitant ESWT would be administered in tandem at a bone marrow donor site, such as the iliac crest, to precondition resident bone marrow stromal cells (BMSCs) in vivo, priming resident MSCs by enlarging and conditioning their population prior to bone marrow aspiration. The resulting sample could then be treated to further augment cell concentration and injected, under fluoroscopic control, into the non-union site through a percutaneous approach.
Collapse
Affiliation(s)
- Valerio Sansone
- Department of Orthopaedics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy; Galeazzi Orthopaedic Institute, Via Riccardo Galeazzi, 4, 20161 Milan, Italy.
| | - Manuel Brañes
- Universidad de Chile, Facultad de Ciencias, Las Palmeras 3425, Ñuñoa, Región Metropolitana, Chile
| | - Pietro Romeo
- Galeazzi Orthopaedic Institute, Via Riccardo Galeazzi, 4, 20161 Milan, Italy
| |
Collapse
|
25
|
Desando G, Giavaresi G, Cavallo C, Bartolotti I, Sartoni F, Nicoli Aldini N, Martini L, Parrilli A, Mariani E, Fini M, Grigolo B. Autologous Bone Marrow Concentrate in a Sheep Model of Osteoarthritis: New Perspectives for Cartilage and Meniscus Repair. Tissue Eng Part C Methods 2017; 22:608-19. [PMID: 27151837 DOI: 10.1089/ten.tec.2016.0033] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Cell-based therapies are becoming a valuable tool to treat osteoarthritis (OA). This study investigated and compared the regenerative potential of bone marrow concentrate (BMC) and mesenchymal stem cells (MSC), both engineered with Hyaff(®)-11 (HA) for OA treatment in a sheep model. METHODS OA was induced via unilateral medial meniscectomy. Bone marrow was aspirated from the iliac crest, followed by concentration processes or cell isolation and expansion to obtain BMC and MSC, respectively. Treatments consisted of autologous BMC and MSC seeded onto HA. The regenerative potential of bone, cartilage, menisci, and synovia was monitored using macroscopy, histology, immunohistochemistry, and micro-computed tomography at 12 weeks post-op. Data were analyzed using the general linear model with adjusted Sidak's multiple comparison and Spearman's tests. RESULTS BMC-HA treatment showed a greater repair ability in inhibiting OA progression compared to MSC-HA, leading to a reduction of inflammation in cartilage, meniscus, and synovium. Indeed, the decrease of inflammation positively contributed to counteract the progression of fibrotic and hypertrophic processes, known to be involved in tissue failure. Moreover, the treatment with BMC-HA showed the best results in allowing meniscus regeneration. Minor healing effects were noticed at bone level for both cell strategies; however, a downregulation of subchondral bone thickness (Cs.Th) was found in both cell treatments compared to the OA group in the femur. CONCLUSION The transplantation of BMC-HA provided the best effects in supporting regenerative processes in cartilage, meniscus, and synovium and at less extent in bone. On the whole, both MSC and BMC combined with HA reduced inflammation and contributed to switch off fibrotic and hypertrophic processes. The observed regenerative potential by BMC-HA on meniscus could open new perspectives, suggesting its use not only for OA care but also for the treatment of meniscal lesions, even if further analyses are necessary to confirm its healing potential at long-term follow-up.
Collapse
Affiliation(s)
- Giovanna Desando
- 1 Laboratory RAMSES, Rizzoli Orthopedic Institute , Bologna, Italy
| | - Gianluca Giavaresi
- 2 Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute , Bologna, Italy .,3 Laboratory BITTA, Rizzoli Orthopedic Institute , Bologna, Italy
| | - Carola Cavallo
- 1 Laboratory RAMSES, Rizzoli Orthopedic Institute , Bologna, Italy
| | - Isabella Bartolotti
- 4 Laboratory of Immunorheumatology and Tissue Regeneration, Rizzoli Orthopedic Institute , Bologna, Italy
| | - Federica Sartoni
- 1 Laboratory RAMSES, Rizzoli Orthopedic Institute , Bologna, Italy
| | - Nicolò Nicoli Aldini
- 2 Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute , Bologna, Italy .,3 Laboratory BITTA, Rizzoli Orthopedic Institute , Bologna, Italy
| | - Lucia Martini
- 2 Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute , Bologna, Italy .,3 Laboratory BITTA, Rizzoli Orthopedic Institute , Bologna, Italy
| | | | - Erminia Mariani
- 4 Laboratory of Immunorheumatology and Tissue Regeneration, Rizzoli Orthopedic Institute , Bologna, Italy .,5 Department of Medical and Surgical Science, University of Bologna , Bologna, Italy
| | - Milena Fini
- 2 Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute , Bologna, Italy .,3 Laboratory BITTA, Rizzoli Orthopedic Institute , Bologna, Italy
| | - Brunella Grigolo
- 1 Laboratory RAMSES, Rizzoli Orthopedic Institute , Bologna, Italy .,4 Laboratory of Immunorheumatology and Tissue Regeneration, Rizzoli Orthopedic Institute , Bologna, Italy
| |
Collapse
|
26
|
Differentiation of Human Amniotic Mesenchymal Stem Cells into Human Anterior Cruciate Ligament Fibroblast Cells by In Vitro Coculture. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7360354. [PMID: 29085840 PMCID: PMC5632453 DOI: 10.1155/2017/7360354] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/13/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Anterior cruciate ligament injuries are common in humans, though cellular components of the knee have little regenerative or proliferation potential. This study investigated the differentiation of human amnion-derived mesenchymal stem cells (hAMSCs) into human anterior cruciate ligament fibroblasts (hACLFs) in vitro through induction with bFGF and TGF-β1 with coculture systems. Groups A and B comprised hAMSCs at the 3rd passage cultured with and without bFGF and TGF-β1, respectively; Groups C and D consisted of hAMSCs and hACLFs in monolayer coculture with and without bFGF and TGF-β1, respectively; Groups E and F were composed of hAMSCs and hACLFs in Transwell coculture with and without bFGF and TGF-β1, respectively. Cell morphology and proliferation were recorded. Protein expression and relative mRNA expression were evaluated in each group. Cell proliferation was significantly higher in the induced groups than in the noninduced groups. Protein expression increased over time with the highest expression observed in Group E. mRNA levels were significantly higher in Group E than in other groups. This study is the first to demonstrate the use of the Transwell coculture system for this purpose, and hAMSCs were successfully differentiated into hACLFs. Thus, hAMSCs may be a superior choice for hACLF differentiation via Transwell coculture.
Collapse
|
27
|
Carballo CB, Lebaschi A, Rodeo SA. Cell-based approaches for augmentation of tendon repair. TECHNIQUES IN SHOULDER & ELBOW SURGERY 2017; 18:e6-e14. [PMID: 29276433 PMCID: PMC5737795 DOI: 10.1097/bte.0000000000000132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-based approaches are among the principal interventions in orthobiologics to improve tendon and ligament healing and to combat degenerative processes. The number of options available for investigation are expanding rapidly and investigators have an increasing number of cell types to choose from for research purposes. However, in part due to the current regulatory environment, the list of available cells at clinicians' disposal for therapeutic purposes is still rather limited. In this review, we present an overview of the main cellular categories in current use. Notable recent developments in cell-based approaches include the introduction of diverse sources of mesenchymal stem cells, pluripotent cells of extra-embryonic origin, and the emerging popularity of fully differentiated cells such as tenocytes and endothelial cells. Delivery strategies are discussed and a succinct discussion of the current regulatory environment in the United States is presented.
Collapse
Affiliation(s)
- Camila B Carballo
- Laboratory for Joint Tissue Repair and Regeneration, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery
| | - Amir Lebaschi
- Laboratory for Joint Tissue Repair and Regeneration, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery
| | - Scott A Rodeo
- Laboratory for Joint Tissue Repair and Regeneration, Orthopedic Soft Tissue Research Program, Hospital for Special Surgery
| |
Collapse
|
28
|
Zou G, Li Y, Jin Y, Zhu X, Yang J, Wang S, You Q, Xiong H, Liu Y. [ In vitrodifferentiation of human amniotic mesenchymal stem cells into ligament fibroblasts after induced by transforming growth factor β 1 and vascular endothelial growth factor]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:582-593. [PMID: 29798549 PMCID: PMC8498243 DOI: 10.7507/1002-1892.201612090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/07/2017] [Indexed: 11/03/2022]
Abstract
Objective To investigate whether human amniotic mesenchymal stem cells (hAMSCs) have the characteristics of mesenchymal stem cells (MSCs) and the differentiation capacity into ligament fibroblasts in vitro. Methods The hAMSCs were separated through trypsin and collagenase digestion from placenta, the phenotypic characteristics of hAMSCs were detected by flow cytometry, the cytokeratin-19 (CK-19) and vimentin expression of hAMSCs were tested through immunofluorescence staining. The hAMSCs at the 3rd passage were cultured with L-DMEM/F12 medium containing transforming growth factor β 1 (TGF-β 1) and vascular endothelial growth factor (VEGF) as the experimental group and with single L-DMEM/F12 medium as the control group. The morphology of hAMSCs was observed by inverted phase contrast microscope; the cellular activities and ability of proliferation were examined by cell counting kit-8 (CCK-8) method; the ligament fibroblasts related protein expressions including collagen type I, collagen type III, Fibronectin, and Tenascin-C were detected by immunofluorescence staining; specific mRNA expressions of ligament fibroblasts and angiogenesis including collagen type I, collagen type III, Fibronectin, α-smooth muscle actin (α-SMA), and VEGF were measured by real-time fluorescence quantitative PCR. Results The hAMSCs presented monolayer and adherent growth under inverted phase contrast microscope; the flow cytometry results demonstrated that hAMSCs expressed the MSCs phenotypes; the immunofluorescence staining results indicated the hAMSCs had high expression of the vimentin and low expression of CK-19; the hAMSCs possessed the differentiation ability into the osteoblasts, chondroblasts, and lipoblasts. The CCK-8 results displayed that cells reached the peak of growth curve at 7 days in each group, and the proliferation ability in the experimental group was significantly higher than that in the control group at 7 days ( P<0.05). The immunofluorescence staining results showed that the expressions of collagen type I, collagen type III, Fibronectin, and Tenascin-C in the experimental group were significantly higher than those in the control group at 5, 10, and15 days after culture ( P<0.05). The real-time fluorescence quantitative PCR results revealed that the mRNA relative expressions had an increasing tendency at varying degrees with time in the experimental group ( P<0.05). The relative mRNA expressions of collagen type I, collagen type III, Fibronectin, α-SMA, and VEGF in the experimental group were significantly higher than those in the control group at the other time points ( P<0.05), but no significant difference was found in the relative mRNA expressions of collagen type I, collagen type III, and VEGF between 2 groups at 5 days ( P>0.05). Conclusion The hAMSCs possesses the characteristics of MSCs and good proliferation ability which could be chosen as seed cell source in tissue engineering. The expressions of ligament fibroblasts and angiogenesis related genes could be up-regulated, after induction in vitro, and the synthesis of ligament fibroblasts related proteins could be strengthened. In addition, the application of TGF-β 1 and VEGF could be used as growth factors sources in constructing tissue engineered ligament.
Collapse
Affiliation(s)
- Gang Zou
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | | | - Ying Jin
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Xizhong Zhu
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Jibin Yang
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Shengmin Wang
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Qi You
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Huazhang Xiong
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Yi Liu
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000,
| |
Collapse
|
29
|
Platelet-rich Plasma and Bone Marrow-derived Mesenchymal Stem Cells in Sports Medicine. Sports Med Arthrosc Rev 2017; 24:69-73. [PMID: 27135289 DOI: 10.1097/jsa.0000000000000105] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative medicine is a fast-growing field in orthopedic sports medicine. Platelet-rich plasma contains multiple factors that have been shown to augment healing, thereby stimulating its use in multiple areas of acute and chronic injuries. Mesenchymal stem cells have pluripotent potential to form into tissues pertinent to orthopedics, such as cartilage and bone. As such, there is been a surge in the research directed toward steering those stem cells into a particular lineage as part of treatment for a variety of soft-tissue, cartilage, and bone pathologies. Overall, there are promising reports of their potential success, but there is a need for continued investigation into the efficacy of platelet-rich plasma and stem cells in sports medicine.
Collapse
|
30
|
Anz AW, Branch EA, Rodriguez J, Chillemi F, Bruce JR, Murphy MB, Suzuki RK, Andrews JR. Viable Stem Cells Are in the Injury Effusion Fluid and Arthroscopic Byproducts From Knee Cruciate Ligament Surgery: An In Vivo Analysis. Arthroscopy 2017; 33:790-797. [PMID: 28043750 DOI: 10.1016/j.arthro.2016.09.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/21/2016] [Accepted: 09/28/2016] [Indexed: 02/02/2023]
Abstract
PURPOSE To examine the number of viable stem cells contained in the postinjury effusion fluid and the waste byproducts of arthroscopic cruciate ligament surgery. METHODS This study included patients older than 18 years of age with acute (<5 weeks old) cruciate ligament injuries requiring arthroscopic surgery. The postinjury effusion fluid (effusion fluid), fat pad and cruciate ligament stump debridement tissue (byproduct tissue), and arthroscopic fluid collected during fat pad and/or stump debridement (byproduct fluid) were collected at the time of surgery from 30 individuals. Specimens were analyzed, investigating cell viability, nucleated cell counts, cell concentrations, colony-forming unit assays, and flow cytometry. Samples from the first 20 individuals were collected in small specimen containers, and samples from the last 10 individuals were collected in larger specimen containers. RESULTS Cells of the injury effusion exhibited the greatest viability (86.4 ± 1.31%) when compared with the small volume harvest byproduct tissue (50.2 ± 2.5%, P = .0001), small volume harvest byproduct fluid (48.8 ± 1.88%, P = .0001), large volume harvest byproduct tissue (70.1 ± 5.6%, P = .0001), and large volume harvest byproduct fluid (60.3 ± 3.41%, P = .0001). The culture analysis of fibroblast colony-forming units found on average 1916 ± 281 progenitor cells in the effusion fluid, 2488 ± 778 progenitor cells in the byproduct tissue, and 2357 ± 339 progenitor cells in the byproduct fluid. Flow cytometry confirmed the presence of immature cells and the presence of cells with markers typically expressed by known stem cell populations. CONCLUSIONS Viable stem cells are mobilized to the postinjury effusion at the time of cruciate ligament injury and can be found in the byproduct waste of cruciate ligament surgery. CLINICAL RELEVANCE The methodology around effusion fluid and byproduct tissue capture during cruciate ligament surgery should be investigated further. Cell amounts available from these tissues with current technologies are not sufficient for immediate evidence-based clinical application.
Collapse
Affiliation(s)
- Adam W Anz
- Andrews Institute for Orthopaedics & Sports Medicine, Gulf Breeze, Florida, U.S.A..
| | - Eric A Branch
- Andrews Institute for Orthopaedics & Sports Medicine, Gulf Breeze, Florida, U.S.A
| | - John Rodriguez
- Andrews Institute for Orthopaedics & Sports Medicine, Gulf Breeze, Florida, U.S.A
| | - Fellipo Chillemi
- Andrews Institute for Orthopaedics & Sports Medicine, Gulf Breeze, Florida, U.S.A
| | - Jeremy R Bruce
- Andrews Institute for Orthopaedics & Sports Medicine, Gulf Breeze, Florida, U.S.A
| | | | | | - James R Andrews
- Andrews Institute for Orthopaedics & Sports Medicine, Gulf Breeze, Florida, U.S.A
| |
Collapse
|
31
|
Ćuti T, Antunović M, Marijanović I, Ivković A, Vukasović A, Matić I, Pećina M, Hudetz D. Capacity of muscle derived stem cells and pericytes to promote tendon graft integration and ligamentization following anterior cruciate ligament reconstruction. INTERNATIONAL ORTHOPAEDICS 2017; 41:1189-1198. [PMID: 28299448 DOI: 10.1007/s00264-017-3437-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/01/2017] [Indexed: 01/08/2023]
Abstract
PURPOSE The aim of this study is to examine the capacity of muscle tissue preserved on hamstring tendons forming candy-stripe grafts in order to improve tendon to bone ingrowth and ligamentization. We hypothesized that muscle tissue does possess a stem cell population that could enhance the healing process of the ACL graft when preserved on the tendons. METHODS Human samples from gracilis and semitendinosus muscles were collected during ACL surgery from ten patients and from these tissue samples human muscle-derived stem cells and tendon-derived stem cells were isolated and propagated. Both stem cell populations were in-vitro differentiated into osteogenic lineage. Alkaline phosphatase activity was determined at days zero and 14 of the osteogenic induction and von Kossa staining to assess mineralization of the cultures. Total RNA was collected from osteoblast cultures and real time quantitative PCR was performed. Western-blot for osteocalcin and collagen type I followed protein isolation. Immunofluorescence double labeling of pericytes in muscle and tendon tissue was performed. RESULTS Mesenchymal stem cells from muscle and tendon tissue were isolated and expanded in cell culture. More time was needed to grow the tendon derived culture compared to muscle derived culture. Muscle derived stem cells exhibited more alkaline phosphatase actvity compared to tendon derived stem cells, whereas tendon derived stem cells formed more mineralized nodules after 14 days of osteoinduction. Muscle derived stem cells exhibited higher expression levels of bone sialoprotein, and tendon derived stem cells showed higher expression of dental-matrix-protein 1 and osteocalcin. Immunofluorescent staining against pericytes indicated that they are more abundant in muscle tissue. CONCLUSIONS These results indicate that muscle tissue is a better source of stem cells than tendon tissue. Achievement of this study is proof that there is vast innate capacity of muscle tissue for enhancement of bone-tendon integration and ligamentization of ACL hamstring grafts and consequently muscle tissue should not be treated as waste after harvesting.
Collapse
Affiliation(s)
- Tomislav Ćuti
- Clinic for Trauma Surgery, University Hospital Center "Sestre Milosrdnice", Vinogradska cesta 29, Zagreb, Croatia
| | - Maja Antunović
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb, Croatia
| | - Inga Marijanović
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb, Croatia
| | - Alan Ivković
- Department for Orthopaedic Surgery, University Hospital "Sveti Duh", Sveti Duh 64, Zagreb, Croatia.,Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, Rijeka, Croatia.,Department of Histology and Embriology, School of Medicine, University of Zagreb, Šalata 3, Zagreb, Croatia
| | - Andreja Vukasović
- Department of Histology and Embriology, School of Medicine, University of Zagreb, Šalata 3, Zagreb, Croatia
| | - Igor Matić
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb, Croatia
| | - Marko Pećina
- Department of Orthopaedic Surgery, School of Medicine University of Zagreb, Šalata 7, Zagreb, Croatia
| | - Damir Hudetz
- Department for Orthopaedic Surgery, University Hospital "Sveti Duh", Sveti Duh 64, Zagreb, Croatia. .,St.Catherine Specialty Hospital, Bračak 8, Zabok, Croatia. .,University of Osijek, Medical School, Osijek, Croatia.
| |
Collapse
|
32
|
Farah S, Anderson DG, Langer R. Physical and mechanical properties of PLA, and their functions in widespread applications - A comprehensive review. Adv Drug Deliv Rev 2016; 107:367-392. [PMID: 27356150 DOI: 10.1016/j.addr.2016.06.012] [Citation(s) in RCA: 1090] [Impact Index Per Article: 136.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 12/28/2022]
Abstract
Poly(lactic acid) (PLA), so far, is the most extensively researched and utilized biodegradable aliphatic polyester in human history. Due to its merits, PLA is a leading biomaterial for numerous applications in medicine as well as in industry replacing conventional petrochemical-based polymers. The main purpose of this review is to elaborate the mechanical and physical properties that affect its stability, processability, degradation, PLA-other polymers immiscibility, aging and recyclability, and therefore its potential suitability to fulfill specific application requirements. This review also summarizes variations in these properties during PLA processing (i.e. thermal degradation and recyclability), biodegradation, packaging and sterilization, and aging (i.e. weathering and hygrothermal). In addition, we discuss up-to-date strategies for PLA properties improvements including components and plasticizer blending, nucleation agent addition, and PLA modifications and nanoformulations. Incorporating better understanding of the role of these properties with available improvement strategies is the key for successful utilization of PLA and its copolymers/composites/blends to maximize their fit with worldwide application needs.
Collapse
|
33
|
The Role of Autophagy in the Maintenance of Stemness and Differentiation of Mesenchymal Stem Cells. Stem Cell Rev Rep 2016; 12:621-633. [DOI: 10.1007/s12015-016-9690-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
34
|
Use of FGF-2 and FGF-18 to direct bone marrow stromal stem cells to chondrogenic and osteogenic lineages. Future Sci OA 2016; 2:FSO142. [PMID: 28116125 PMCID: PMC5242207 DOI: 10.4155/fsoa-2016-0034] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/29/2016] [Indexed: 02/07/2023] Open
Abstract
Aim: Intervertebral disc degeneration/low back pain is the number one global musculoskeletal condition in terms of disability and socioeconomic impact. Materials & methods Multipotent mesenchymal stem cells (MSCs) were cultured in micromass pellets ± FGF-2 or -18 up to 41 days, matrix components were immunolocalized and gene expression monitored by quantitative-reverse transcription PCR. Results: Chondrogenesis occurred earlier in FGF-18 than FGF-2 cultures. Lower COL2A1, COL10A1 and ACAN expression by day 41 indicated a downregulation in chondrocyte hypertrophy. MEF2c, ALPL, were upregulated; calcium, decorin and biglycan, and 4C3 and 7D4 chondroitin sulphate sulfation motifs were evident in FGF-18 but not FGF-2 pellets. Conclusion: FGF-2 and -18 preconditioned MSCs produced cell lineages which promoted chondrogenesis and osteogenesis and may be useful in the production of MSC lineages suitable for repair of cartilaginous tissue defects. Intervertebral disc degeneration and low back pain is the number one global musculoskeletal disorder effecting 80% of the general population. A remedy for this condition is being eagerly sought as part of a WHO research priority. Stem cells are one potential therapy that shows promise in animal models, laboratory studies, and preclinical and early clinical trials. Conditioning of stem cells in the laboratory before injection may improve their efficacy for the alleviation of low back pain. In the present study we have developed a means of improving how stem cells form cartilage and bone, which should be of application in the repair of spinal defects.
Collapse
|
35
|
Chen YH, Chung CC, Liu YC, Yeh SP, Hsu JL, Hung MC, Su HL, Li LY. Enhancer of Zeste Homolog 2 and Histone Deacetylase 9c Regulate Age-Dependent Mesenchymal Stem Cell Differentiation into Osteoblasts and Adipocytes. Stem Cells 2016; 34:2183-93. [PMID: 27250566 DOI: 10.1002/stem.2400] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 04/18/2016] [Accepted: 04/18/2016] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent precursors that can undergo multilineage differentiation, including osteogenesis and adipogenesis, which are two mutually exclusive events. Previously, we demonstrated that enhancer of zeste homolog 2 (EZH2), the catalytic component of the Polycomb-repressive complex 2, mediates epigenetic silencing of histone deacetylase 9c (HDAC9c) in adipocytes but not in osteoblasts and that HDAC9c accelerates osteogenesis while attenuating adipogenesis of MSCs through inactivation of peroxisome proliferator-activated receptor gamma 2 activity. Importantly, disrupting the balance between adipogenesis and osteogenesis can lead to age-associated bone loss (osteoporosis) and obesity. Here, we investigated the relationship between age, and osteogenic and adipogenic differentiation potential of MSCs by comparing EZH2 and HDAC9c expression in osteoblasts and adipocytes of both human and mice origins to determine whether the EZH2-HDAC9c axis regulates age-associated osteoporosis and obesity. Our findings indicated that a decline in HDAC9c expression over time was accompanied by increased EZH2 expression and suggested that a therapeutic intervention for age-associated osteoporosis and obesity may be feasible by targeting the EZH2-HDAC9c axis. Stem Cells 2016;34:2183-2193.
Collapse
Affiliation(s)
- Ya-Huey Chen
- Graduate Institute of Cancer Biology, College of Medicine, Taichung, Taiwan.,Cancer Biology and Drug Discovery Ph.D. Program, College of Medicine, China Medical University, Taichung, Taiwan.,Center for Molecular Medicine, Taichung, Taiwan
| | | | - Yu-Chia Liu
- Graduate Institute of Cancer Biology, College of Medicine, Taichung, Taiwan
| | - Su-Peng Yeh
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, the University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Mien-Chie Hung
- Graduate Institute of Cancer Biology, College of Medicine, Taichung, Taiwan.,Center for Molecular Medicine, Taichung, Taiwan.,Department of Molecular and Cellular Oncology, the University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.,Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Long-Yuan Li
- Graduate Institute of Cancer Biology, College of Medicine, Taichung, Taiwan.,Center for Molecular Medicine, Taichung, Taiwan.,Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
36
|
Hayes AJ, Hughes CE, Smith SM, Caterson B, Little CB, Melrose J. The CS Sulfation Motifs 4C3, 7D4, 3B3[-]; and Perlecan Identify Stem Cell Populations and Their Niches, Activated Progenitor Cells and Transitional Areas of Tissue Development in the Fetal Human Elbow. Stem Cells Dev 2016; 25:836-47. [PMID: 27068010 DOI: 10.1089/scd.2016.0054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We compared the immunohistochemical distribution of (1) the novel chondroitin sulfate (CS) sulfation motifs 7D4, 4C3, and 3B3[-], (2) native heparan sulfate (HS) and Δ-HS "stubs" generated by heparitinase III digestion and (3) the HS-proteoglycan (PG), perlecan, in the fetal human elbow joint. Putative stem cell populations associated with hair bulbs, humeral perichondrium, humeral and ulnar rudiment stromal/perivascular tissues expressed the CS motifs 4C3, 7D4, and 3B3[-] along with perlecan in close association but not colocalized. Chondrocytes in the presumptive articular cartilage of the fetal elbow expressed the 4C3 and 7D4 CS sulfation motifs consistent with earlier studies on the expression of these motifs in knee cartilage following joint cavitation. This study also indicated that hair bulbs, skin, perichondrium, and rudiment stroma were all perlecan-rich progenitor cell niches that contributed to the organization and development of the human fetal elbow joint and associated connective tissues. One of the difficulties in determining the precise role of stem cells in tissue development and repair processes is their short engraftment period and the lack of specific markers, which differentiate the activated stem cell lineages from the resident cells. The CS sulfation motifs 7D4, 4C3, and 3B3[-] decorate cell surface PGs on activated stem/progenitor cells and thus can be used to identify these cells in transitional areas of tissue development and in repair tissues and may be applicable to determining a more precise mode of action of stem cells in these processes. Isolation of perlecan from 12 to 14 week gestational age fetal knee rudiments demonstrated that perlecan in these fetal tissues was a HS-CS hybrid PG further supporting roles for CS in tissue development.
Collapse
Affiliation(s)
- Anthony J Hayes
- 1 Bioimaging Unit, Cardiff School of Biosciences, University of Cardiff , United Kingdom
| | - Clare E Hughes
- 2 School of Biosciences, University of Cardiff , Cardiff, United Kingdom
| | - Susan M Smith
- 3 Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney , St. Leonards, New South Wales, Australia
| | - Bruce Caterson
- 2 School of Biosciences, University of Cardiff , Cardiff, United Kingdom
| | - Christopher B Little
- 3 Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney , St. Leonards, New South Wales, Australia .,4 Sydney Medical School, Northern, The University of Sydney , Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| | - James Melrose
- 3 Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney , St. Leonards, New South Wales, Australia .,4 Sydney Medical School, Northern, The University of Sydney , Royal North Shore Hospital, St. Leonards, New South Wales, Australia .,5 Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales , Sydney, New South Wales, Australia
| |
Collapse
|
37
|
Filardo G, Perdisa F, Roffi A, Marcacci M, Kon E. Stem cells in articular cartilage regeneration. J Orthop Surg Res 2016; 11:42. [PMID: 27072345 PMCID: PMC4830073 DOI: 10.1186/s13018-016-0378-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising option to treat articular defects and early osteoarthritis (OA) stages. However, both their potential and limitations for a clinical use remain controversial. Thus, the aim of this systematic review was to examine MSCs treatment strategies in clinical settings, in order to summarize the current evidence of their efficacy for the treatment of cartilage lesions and OA.Among the 60 selected studies, 7 were randomized, 13 comparative, 31 case series, and 9 case reports; 26 studies reported the results after injective administration, whereas 33 used surgical implantation. One study compared the two different modalities. With regard to the cell source, 20 studies concerned BMSCs, 17 ADSCs, 16 BMC, 5 PBSCs, 1 SDSCs, and 1 compared BMC versus PBSCs. Overall, despite the increasing literature on this topic, the evidence is still limited, in particular for high-level studies. On the other hand, the available studies allow to draw some indications. First, no major adverse events related to the treatment or to the cell harvest have been reported. Second, a clinical benefit of using MSCs therapies has been reported in most of the studies, regardless of cell source, indication, or administration method. This effectiveness has been reflected by clinical improvements and also positive MRI and macroscopic findings, whereas histologic features gave more controversial results among different studies. Third, young age, lower BMI, smaller lesion size for focal lesions, and earlier stages of OA joints have been shown to correlate with better outcomes, even though the available data strength does not allow to define clear cutoff values. Finally, definite trends can be observed with regard to the delivery method: currently cultured cells are mostly being administered by i.a. injection, while one-step surgical implantation is preferred for cell concentrates. In conclusion, while promising results have been shown, the potential of these treatments should be confirmed by reliable clinical data through double-blind, controlled, prospective and multicenter studies with longer follow-up, and specific studies should be designed to identify the best cell sources, manipulation, and delivery techniques, as well as pathology and disease phase indications.
Collapse
Affiliation(s)
- Giuseppe Filardo
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Francesco Perdisa
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Alice Roffi
- />Nanobiotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Maurilio Marcacci
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
- />Nanobiotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Elizaveta Kon
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
- />Nanobiotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
38
|
Binder BYK, Sagun JE, Leach JK. Reduced serum and hypoxic culture conditions enhance the osteogenic potential of human mesenchymal stem cells. Stem Cell Rev Rep 2016; 11:387-93. [PMID: 25173881 DOI: 10.1007/s12015-014-9555-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
UNLABELLED Current protocols for inducing osteogenic differentiation in mesenchymal stem/stromal cells (MSCs) in culture for tissue engineering applications depend on the use of biochemical supplements. However, standard in vitro culture conditions expose cells to ambient oxygen concentrations and high levels of serum (21% O2, 10% FBS) that do not accurately recapitulate the physiological milieu. While we and others have examined MSC behavior under hypoxia, the synergistic effect of low serum levels, such as those present in ischemic injury sites, on osteogenic differentiation has not been clearly examined. We hypothesized that a concomitant reduction of serum and O2 would enhance in vitro osteogenic differentiation of MSCs by more accurately mimicking the fracture microenvironment. We show that serum deprivation, in conjunction with hypoxia, potentiates osteogenic differentiation in MSCs. These data demonstrate the role of serum levels in regulating osteogenesis and its importance in optimizing MSC differentiation strategies. HIGHLIGHTS Serum levels, in addition to hypoxia, have a significant effect on MSC osteogenic differentiation. Both naïve and osteogenically induced MSCs exhibit higher osteogenic markers in reduced serum. MSCs deposit the most calcium under 5% O2 in osteogenic media supplemented with 5% FBS. Standard culture conditions (21% O2, 10% FBS) may not be optimal for MSC osteogenic differentiation.
Collapse
Affiliation(s)
- Bernard Y K Binder
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Drive, Davis, CA, 95616, USA
| | | | | |
Collapse
|
39
|
Gohil SV, Kuo C, Adams DJ, Maye P, Rowe DW, Nair LS. Evaluation of the donor cell contribution in rh
BMP
‐2 mediated bone formation with chitosan thermogels using fluorescent protein reporter mice. J Biomed Mater Res A 2016; 104:928-41. [DOI: 10.1002/jbm.a.35634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/18/2015] [Accepted: 12/18/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Shalini V. Gohil
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
| | - Chia‐Ling Kuo
- Connecticut Institute for Clinical and Translational Science, Institute for Systems Genomics, University of ConnecticutFarmington Connecticut06030
| | - Douglas J. Adams
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
| | - Peter Maye
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - David W. Rowe
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - Lakshmi S. Nair
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
- Departments of Material Science and Engineering, Biomedical Engineering and Institute of Material ScienceUniversity of ConnecticutStorrs Connecticut06269
| |
Collapse
|
40
|
Sheng G. The developmental basis of mesenchymal stem/stromal cells (MSCs). BMC DEVELOPMENTAL BIOLOGY 2015; 15:44. [PMID: 26589542 PMCID: PMC4654913 DOI: 10.1186/s12861-015-0094-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mesenchymal Stem/Stromal Cells (MSCs) define a population of progenitor cells capable of giving rises to at least three mesodermal lineages in vitro, the chondrocytes, osteoblasts and adipocytes. The validity of MSCs in vivo has been questioned because their existence, either as a homogeneous progenitor cell population or as a stem cell lineage, has been difficult to prove. The wide use of primary MSCs in regenerative and therapeutic applications raises ethical and regulatory concerns in many countries. In contrast to hematopoietic stem cells, a parallel concept which carries an embryological emphasis from its outset, MSCs have attracted little interest among developmental biologists and the embryological basis for their existence, or lack thereof, has not been carefully evaluated. METHODS This article provides a brief, embryological overview of these three mesoderm cell lineages and offers a framework of ontological rationales for the potential existence of MSCs in vivo. RESULTS Emphasis is given to the common somatic lateral plate mesoderm origin of the majority of body's adipose and skeletal tissues and of the major sources used for MSC derivation clinically. Support for the MSC hypothesis also comes from a large body of molecular and lineage analysis data in vivo. CONCLUSIONS It is concluded that despite the lack of a definitive proof, the MSC concept has a firm embryological basis and that advances in MSC research can be facilitated by achieving a better integration with developmental biology.
Collapse
Affiliation(s)
- Guojun Sheng
- Sheng Laboratory, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.
| |
Collapse
|
41
|
Agarwal R, García AJ. Biomaterial strategies for engineering implants for enhanced osseointegration and bone repair. Adv Drug Deliv Rev 2015; 94:53-62. [PMID: 25861724 DOI: 10.1016/j.addr.2015.03.013] [Citation(s) in RCA: 421] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 02/08/2015] [Accepted: 03/17/2015] [Indexed: 12/11/2022]
Abstract
Bone tissue has a remarkable ability to regenerate and heal itself. However, large bone defects and complex fractures still present a significant challenge to the medical community. Current treatments center on metal implants for structural and mechanical support and auto- or allo-grafts to substitute long bone defects. Metal implants are associated with several complications such as implant loosening and infections. Bone grafts suffer from donor site morbidity, reduced bioactivity, and risk of pathogen transmission. Surgical implants can be modified to provide vital biological cues, growth factors and cells in order to improve osseointegration and repair of bone defects. Here we review strategies and technologies to engineer metal surfaces to promote osseointegration with the host tissue. We also discuss strategies for modifying implants for cell adhesion and bone growth via integrin signaling and growth factor and cytokine delivery for bone defect repair.
Collapse
|
42
|
Zigdon-Giladi H, Rudich U, Michaeli Geller G, Evron A. Recent advances in bone regeneration using adult stem cells. World J Stem Cells 2015; 7:630-640. [PMID: 25914769 PMCID: PMC4404397 DOI: 10.4252/wjsc.v7.i3.630] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/30/2014] [Accepted: 01/20/2015] [Indexed: 02/06/2023] Open
Abstract
Bone is a highly vascularized tissue reliant on the close spatial and temporal association between blood vessels and bone cells. Therefore, cells that participate in vasculogenesis and osteogenesis play a pivotal role in bone formation during prenatal and postnatal periods. Nevertheless, spontaneous healing of bone fracture is occasionally impaired due to insufficient blood and cellular supply to the site of injury. In these cases, bone regeneration process is interrupted, which might result in delayed union or even nonunion of the fracture. Nonunion fracture is difficult to treat and have a high financial impact. In the last decade, numerous technological advancements in bone tissue engineering and cell-therapy opened new horizon in the field of bone regeneration. This review starts with presentation of the biological processes involved in bone development, bone remodeling, fracture healing process and the microenvironment at bone healing sites. Then, we discuss the rationale for using adult stem cells and listed the characteristics of the available cells for bone regeneration. The mechanism of action and epigenetic regulations for osteogenic differentiation are also described. Finally, we review the literature for translational and clinical trials that investigated the use of adult stem cells (mesenchymal stem cells, endothelial progenitor cells and CD34+ blood progenitors) for bone regeneration.
Collapse
|
43
|
Peric M, Dumic-Cule I, Grcevic D, Matijasic M, Verbanac D, Paul R, Grgurevic L, Trkulja V, Bagi CM, Vukicevic S. The rational use of animal models in the evaluation of novel bone regenerative therapies. Bone 2015; 70:73-86. [PMID: 25029375 DOI: 10.1016/j.bone.2014.07.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/30/2014] [Accepted: 07/05/2014] [Indexed: 12/31/2022]
Abstract
Bone has a high potential for endogenous self-repair. However, due to population aging, human diseases with impaired bone regeneration are on the rise. Current strategies to facilitate bone healing include various biomolecules, cellular therapies, biomaterials and different combinations of these. Animal models for testing novel regenerative therapies remain the gold standard in pre-clinical phases of drug discovery and development. Despite improvements in animal experimentation, excessive poorly designed animal studies with inappropriate endpoints and inaccurate conclusions are being conducted. In this review, we discuss animal models, procedures, methods and technologies used in bone repair studies with the aim to assist investigators in planning and performing scientifically sound experiments that respect the wellbeing of animals. In the process of designing an animal study for bone repair investigators should consider: skeletal characteristics of the selected animal species; a suitable animal model that mimics the intended clinical indication; an appropriate assessment plan with validated methods, markers, timing, endpoints and scoring systems; relevant dosing and statistically pre-justified sample sizes and evaluation methods; synchronization of the study with regulatory requirements and additional evaluations specific to cell-based approaches. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Mihaela Peric
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia.
| | - Ivo Dumic-Cule
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia
| | - Danka Grcevic
- University of Zagreb School of Medicine, Department of Physiology and Immunology, Salata 3, Zagreb, Croatia
| | - Mario Matijasic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia
| | - Donatella Verbanac
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Department for Intercellular Communication, Salata 2, Zagreb, Croatia
| | - Ruth Paul
- Paul Regulatory Services Ltd, Fisher Hill Way, Cardiff CF15 8DR, UK
| | - Lovorka Grgurevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia
| | - Vladimir Trkulja
- University of Zagreb School of Medicine, Department of Pharmacology, Salata 11, Zagreb, Croatia
| | - Cedo M Bagi
- Pfizer Inc., Global Research and Development, Global Science and Technology, 100 Eastern Point Road, Groton, CT 06340, USA
| | - Slobodan Vukicevic
- University of Zagreb School of Medicine, Center for Translational and Clinical Research, Laboratory for Mineralized Tissues, Salata 11, Zagreb, Croatia.
| |
Collapse
|
44
|
Guerrero J, Oliveira H, Catros S, Siadous R, Derkaoui SM, Bareille R, Letourneur D, Amédée J. The use of total human bone marrow fraction in a direct three-dimensional expansion approach for bone tissue engineering applications: focus on angiogenesis and osteogenesis. Tissue Eng Part A 2014; 21:861-74. [PMID: 25333855 DOI: 10.1089/ten.tea.2014.0367] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current approaches in bone tissue engineering have shown limited success, mostly owing to insufficient vascularization of the construct. A common approach consists of co-culture of endothelial cells and osteoblastic cells. This strategy uses cells from different sources and differentiation states, thus increasing the complexity upstream of a clinical application. The source of reparative cells is paramount for the success of bone tissue engineering applications. In this context, stem cells obtained from human bone marrow hold much promise. Here, we analyzed the potential of human whole bone marrow cells directly expanded in a three-dimensional (3D) polymer matrix and focused on the further characterization of this heterogeneous population and on their ability to promote angiogenesis and osteogenesis, both in vitro and in vivo, in a subcutaneous model. Cellular aggregates were formed within 24 h and over the 12-day culture period expressed endothelial and bone-specific markers and a specific junctional protein. Ectopic implantation of the tissue-engineered constructs revealed osteoid tissue and vessel formation both at the periphery and within the implant. This work sheds light on the potential clinical use of human whole bone marrow for bone regeneration strategies, focusing on a simplified approach to develop a direct 3D culture without two-dimensional isolation or expansion.
Collapse
Affiliation(s)
- Julien Guerrero
- 1 Inserm, U1026, Tissue Bioengineering, University of Bordeaux , Bordeaux, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Anderson JA, Little D, Toth AP, Moorman CT, Tucker BS, Ciccotti MG, Guilak F. Stem cell therapies for knee cartilage repair: the current status of preclinical and clinical studies. Am J Sports Med 2014; 42:2253-61. [PMID: 24220016 PMCID: PMC4019709 DOI: 10.1177/0363546513508744] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Articular cartilage damage of the knee is common, causing significant morbidity worldwide. Many adult tissues contain cells that are able to differentiate into multiple cell types, including chondrocytes. These stem cells have gained significant attention over the past decade and may become frontline management for cartilage defects in the very near future. PURPOSE The role of stem cells in the treatment of knee osteochondral defects was reviewed. Recent animal and clinical studies were reviewed to determine the benefits and potential outcomes of using stem cells for cartilage defects. STUDY DESIGN Literature review. METHODS A PubMed search was undertaken. The key phrase "stem cells and knee" was used. The search included reviews and original articles over an unlimited time period. From this search, articles outlining animal and clinical trials were selected. A search of current clinical trials in progress was performed on the clinicaltrials.gov website, and "stem cells and knee" was used as the search phrase. RESULTS Stem cells have been used in many recent in vitro and animal studies. A number of cell-based approaches for cartilage repair have progressed from preclinical animal studies into clinical trials. CONCLUSION The use of stem cells for the treatment of cartilage defects is increasing in animal and clinical studies. Methods of delivery of stem cells to the knee's cartilage vary from direct injection to implantation with scaffolds. While these approaches are highly promising, there is currently limited evidence of a direct clinical benefit, and further research is required to assess the overall outcome of stem cell therapies for knee cartilage repair.
Collapse
Affiliation(s)
- John A. Anderson
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina.
,Rothman Institute Cartilage Center, Rothman Institute, Philadelphia, Pennsylvania.
,Address correspondence to Rothman Institute Cartilage Center, 925 Chestnut Street, Philadelphia, PA 19107 ()
| | - Dianne Little
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
| | - Alison P. Toth
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
| | - Claude T. Moorman
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
| | - Bradford S. Tucker
- Rothman Institute Cartilage Center, Rothman Institute, Philadelphia, Pennsylvania
| | - Michael G. Ciccotti
- Rothman Institute Cartilage Center, Rothman Institute, Philadelphia, Pennsylvania
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
46
|
Hongmin L, Wei Z, Xingrong Y, Jing W, Wenxin G, Jihong C, Xin X, Fulin C. Osteoinductive nanohydroxyapatite bone substitute prepared via in situ hydrothermal transformation of cuttlefish bone. J Biomed Mater Res B Appl Biomater 2014; 103:816-24. [PMID: 25115654 DOI: 10.1002/jbm.b.33261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/22/2014] [Accepted: 07/15/2014] [Indexed: 11/10/2022]
Abstract
The capacity to induce a rapid and controlled healing of bone defects is critical for a bone substitute. Previous studies have reported hydrothermal transformation (HT) of aragonite from cuttlebone (CB) to cuttlebone hydroxyapatite (CBHA). However, the biocompatibility and in vivo characteristic of CBHA have not been fully investigated. We fabricated CBHA via the in situ HT of aragonite from CB. This CBHA exhibited a highly porous structure and nanoscaled surface morphology with a significantly higher protein adsorption rate than CB. Marrow mesenchymal stem cells (MSCs) were seeded and cultured on the CBHA and CB to evaluate their influence on cell proliferation and differentiation. According to scanning electronic microscopy observation and MTT assay, the MSCs adhered and proliferated well on both the CBHA and CB. Compared with the cells on the CB, the MSCs on CBHA exhibited enhanced alkaline phosphatase activity and osteocalcin levels after 13 days of culture. In vivo testing revealed that CBHA could induce ectopic bone formation after implantation, while no bone formation being observed in the CB. These findings demonstrated that a nanoscaled and osteoinductive bone substitute could be produced by hydrothermally transforming an aragonite of CB into a hydroxyapatite.
Collapse
Affiliation(s)
- Li Hongmin
- Rege Lab of Tissue Engineering, Faculty of Life Science, Northwest University, Xi'an, 710075, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Broeckx SY, Maes S, Martinello T, Aerts D, Chiers K, Mariën T, Patruno M, Franco-Obregón A, Spaas JH. Equine Epidermis: A Source of Epithelial-Like Stem/Progenitor Cells with In Vitro and In Vivo Regenerative Capacities. Stem Cells Dev 2014; 23:1134-48. [DOI: 10.1089/scd.2013.0203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sarah Y. Broeckx
- Global Stem cell Technology, Meldert-Lummen, Belgium
- Pell Cell Medicals, Opglabbeek, Belgium
| | | | - Tiziana Martinello
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, Italy
| | | | - Koen Chiers
- Department of Pathology, Bacteriology and Poultry Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Tom Mariën
- Equitom Equine Hospital, Meldert-Lummen, Belgium
| | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, Italy
| | - Alfredo Franco-Obregón
- Department of Biomechanics, Swiss Federal Institute of Technology, ETH, Zürich, Switzerland
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jan H. Spaas
- Global Stem cell Technology, Meldert-Lummen, Belgium
- Pell Cell Medicals, Opglabbeek, Belgium
| |
Collapse
|
48
|
Binder BYK, Genetos DC, Leach JK. Lysophosphatidic acid protects human mesenchymal stromal cells from differentiation-dependent vulnerability to apoptosis. Tissue Eng Part A 2014; 20:1156-64. [PMID: 24131310 DOI: 10.1089/ten.tea.2013.0487] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The survival of transplanted cells and their resulting efficacy in cell-based therapies is markedly impaired due to serum deprivation and hypoxia (SD/H) resulting from poor vascularization within tissue defects. Lysophosphatidic acid (LPA) is a platelet-derived growth factor with pleiotropic effects on many cell types. Mesenchymal stromal cells (MSC) exhibit unique secretory and stimulatory characteristics depending on their differentiation state. In light of the potential of MSC in cell-based therapies, we examined the ability of LPA to abrogate SD/H-induced apoptosis in human MSC at increasing stages of osteogenic differentiation in vitro and assessed MSC survival in vivo. Undifferentiated MSC were rescued from SD/H-induced apoptosis by treatment with both 25 and 100 μM LPA. However, MSC conditioned with osteogenic supplements responded to 25 μM LPA, and cells conditioned with dexamethasone-containing osteogenic media required 100 μM LPA. This rescue was mediated through LPA1 in all cases. The addition of 25 μM LPA enhanced vascular endothelial growth factor (VEGF) secretion by MSC in all conditions, but VEGF availability was not responsible for protection against apoptosis. We also showed that codelivery of 25 μM LPA with MSC in alginate hydrogels significantly improved the persistence of undifferentiated MSC in vivo over 4 weeks as measured by bioluminescence imaging. Osteogenic differentiation alone was protective of SD/H-induced apoptosis in vitro, and the synergistic delivery of LPA did not enhance persistence of osteogenically induced MSC in vivo. These data demonstrate that the capacity of LPA to inhibit SD/H-induced apoptosis in MSC is dependent on both the differentiation state and dosage. This information will be valuable for optimizing osteogenic conditioning regimens for MSC before in vivo implementation.
Collapse
Affiliation(s)
- Bernard Y K Binder
- 1 Department of Biomedical Engineering, University of California , Davis, Davis, California
| | | | | |
Collapse
|
49
|
Application of Biologics in the Treatment of the Rotator Cuff, Meniscus, Cartilage, and Osteoarthritis. J Am Acad Orthop Surg 2014. [DOI: 10.5435/00124635-201402000-00002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2023] Open
|
50
|
Ferris DJ, Frisbie DD, Kisiday JD, McIlwraith CW, Hague BA, Major MD, Schneider RK, Zubrod CJ, Kawcak CE, Goodrich LR. Clinical outcome after intra-articular administration of bone marrow derived mesenchymal stem cells in 33 horses with stifle injury. Vet Surg 2014; 43:255-65. [PMID: 24433318 DOI: 10.1111/j.1532-950x.2014.12100.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 01/01/2013] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To report outcome of horses with femorotibial lesions (meniscal, cartilage or ligamentous) treated with surgery and intra-articular administration of autologous bone marrow derived mesenchymal stem cells (BMSCs). STUDY DESIGN Prospective case series. ANIMALS Horses (n = 33). METHODS Inclusion criteria included horses that had lameness localized to the stifle by diagnostic anesthesia, exploratory stifle arthroscopy and subsequent intra-articular administration of autologous BMSCs. Case details and follow-up were gathered from medical records, owner, trainer or veterinarian. Outcome was defined as returned to previous level of work, returned to work, or failed to return to work. RESULTS Follow-up (mean, 24 months) was obtained; 43% of horses returned to previous level of work, 33% returned to work, and 24% failed to return to work. In horses with meniscal damage (n = 24) a higher percentage in the current study (75%) returned to some level of work compared to those in previous reports (60-63%) that were treated with arthroscopy alone, which resulted in a statistically significant difference between studies (P = .038). Joint flare post injection was reported in 3 horses (9.0%); however, no long-term effects were noted. CONCLUSIONS Intra-articular administration of BMSC postoperatively for stifle lesions appeared to be safe, with morbidity being similar to that of other biologic agents. Improvement in ability to return to work may be realized with BMSC treatment compared to surgery alone in horses with stifle injury.
Collapse
Affiliation(s)
- Dora J Ferris
- Equine Orthopaedic Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | | | | | | | | | | | | | | | | | | |
Collapse
|