1
|
Li Y, Shi G, Liang W, Shang H, Li H, Han Y, Zhao W, Bai L, Qin C. Allogeneic Adipose-Derived Mesenchymal Stem Cell Transplantation Alleviates Atherosclerotic Plaque by Inhibiting Ox-LDL Uptake, Inflammatory Reaction and Endothelial Damage in Rabbits. Cells 2023; 12:1936. [PMID: 37566014 PMCID: PMC10417209 DOI: 10.3390/cells12151936] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of arteries fueled by lipids. It is a major cause of cardiovascular morbidity and mortality. Mesenchymal stem cells have been used for the treatment of atherosclerotic lesions. Adipose-derived stem cells (ADSCs) have been shown to regulate the activation state of macrophages and exhibit anti-inflammatory capabilities. However, the effect of allogeneic ADSCs in the treatment of AS have not been investigated. In this study, the early treatment effect and preliminary mechanism analysis of allogeneic rabbit ADSCs intravenous transplantation were investigated in a high-fat diet rabbit model. The polarization mechanism of rabbit ADSCs on the macrophage was further analyzed in vitro. Compared with the model group, blood lipid levels declined, the plaque area, oxidized low-density lipoprotein (ox-LDL) uptake, scavenger receptor A1 and cluster of differentiation (CD) 36 levels were all significantly reduced, and the accumulation of inflammatory M1 macrophages, apoptosis, interleukin (IL)-6 and tumor necrosis factor (TNF)-α expression were decreased. The endothelial cells (CD31), M2 macrophages, IL-10 and the transforming growth factor (TGF)-β levels increased. In vitro, ADSCs can promote the M1 macrophage phenotypic switch toward the M2 macrophage through their secreted exosomes, and the main mechanism includes increasing arginase 1 expression and IL-10 secretion, declining inducible nitric oxide synthase (iNOS) expression and TNF-α secretion, and activating the STAT6 pathway. Therefore, allogeneic rabbit ADSC transplantation can transmigrate to the aortic atherosclerotic plaques and show a good effect in lowering blood lipids and alleviating atherosclerotic plaque in the early stage of AS by inhibiting ox-LDL uptake, inflammatory response, and endothelial damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chuan Qin
- NHC Key Laboratory of Human Diseases Comparative Medicine, National Human Diseases Animal Model Resource Center, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Science (CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing 100021, China
| |
Collapse
|
2
|
Characterization and functional analysis of the adipose tissue-derived stromal vascular fraction of pediatric patients with osteogenesis imperfecta. Sci Rep 2022; 12:2414. [PMID: 35165317 PMCID: PMC8844034 DOI: 10.1038/s41598-022-06063-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/12/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractPediatric patients with Osteogenesis Imperfecta (OI), a heritable connective tissue disorder, frequently suffer from long bone deformations. Surgical correction often results in bone non-unions, necessitating revision surgery with autogenous bone grafting using bone-marrow-derived stem cells (BM-SC) to regenerate bone. BM-SC harvest is generally invasive and limited in supply; thus, adipose tissue's stromal vascular fraction (SVF) has been introduced as an alternative stem cell reservoir. To elucidate if OI patients' surgical site dissected adipose tissue could be used as autologous bone graft in future, we investigated whether the underlying genetic condition alters SVF's cell populations and in vitro differentiation capacity. After optimizing SVF isolation, we demonstrate successful isolation of SVF of pediatric OI patients and non-OI controls. The number of viable cells was comparable between OI and controls, with about 450,000 per gram tissue. Age, sex, type of OI, disease-causing collagen mutation, or anatomical site of harvest did not affect cell outcome. Further, SVF-containing cell populations were similar between OI and controls, and all isolated SVF's demonstrated chondrogenic, adipogenic, and osteogenic differentiation capacity in vitro. These results indicate that SVF from pediatric OI patients could be used as a source of stem cells for autologous stem cell therapy in OI.
Collapse
|
3
|
The Distraction Osteogenesis Callus: a Review of the Literature. Clin Rev Bone Miner Metab 2022. [DOI: 10.1007/s12018-021-09282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
4
|
Ragni E, Viganò M, De Luca P, Pedrini E, de Girolamo L. Adipose-Derived Stem/Stromal Cells, Stromal Vascular Fraction, and Microfragmented Adipose Tissue. ORTHOBIOLOGICS 2022:47-61. [DOI: 10.1007/978-3-030-84744-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Li Y, Yang Y, Wang M, Zhang X, Bai S, Lu X, Li Y, Waldorff EI, Zhang N, Lee WYW, Li G. High slew rate pulsed electromagnetic field enhances bone consolidation and shortens daily treatment duration in distraction osteogenesis. Bone Joint Res 2021; 10:767-779. [PMID: 34872332 PMCID: PMC8696558 DOI: 10.1302/2046-3758.1012.bjr-2021-0274.r1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Aims Distraction osteogenesis (DO) is a useful orthopaedic procedure employed to lengthen and reshape bones by stimulating bone formation through controlled slow stretching force. Despite its promising applications, difficulties are still encountered. Our previous study demonstrated that pulsed electromagnetic field (PEMF) treatment significantly enhances bone mineralization and neovascularization, suggesting its potential application. The current study compared a new, high slew rate (HSR) PEMF signal, with different treatment durations, with the standard Food and Drug Administration (FDA)-approved signal, to determine if HSR PEMF is a better alternative for bone formation augmentation. Methods The effects of a HSR PEMF signal with three daily treatment durations (0.5, one, and three hours/day) were investigated in an established rat DO model with comparison of an FDA-approved classic signal (three hrs/day). PEMF treatments were applied to the rats daily for 35 days, starting from the distraction phase until termination. Radiography, micro-CT (μCT), biomechanical tests, and histological examinations were employed to evaluate the quality of bone formation. Results All rats tolerated the treatment well and no obvious adverse effects were found. By comparison, the HSR signal (three hrs/day) treatment group achieved the best healing outcome, in that endochondral ossification and bone consolidation were enhanced. In addition, HSR signal treatment (one one hr/day) had similar effects to treatment using the classic signal (three three hrs/day), indicating that treatment duration could be significantly shortened with the HSR signal. Conclusion HSR signal may significantly enhance bone formation and shorten daily treatment duration in DO, making it a potential candidate for a new clinical protocol for patients undergoing DO treatments. Cite this article: Bone Joint Res 2021;10(12):767–779.
Collapse
Affiliation(s)
- Yucong Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yongkang Yang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ming Wang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xiaoting Zhang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Shanshan Bai
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xuan Lu
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yuan Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Erik I Waldorff
- Research & Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Nianli Zhang
- Research & Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| |
Collapse
|
6
|
Pan Q, Li Y, Li Y, Wang H, Kong L, Yang Z, Zhang X, Bai S, Zong Z, Chen G, Lin S, Li G. Local administration of allogeneic or autologous bone marrow-derived mesenchymal stromal cells enhances bone formation similarly in distraction osteogenesis. Cytotherapy 2021; 23:590-598. [PMID: 33546925 DOI: 10.1016/j.jcyt.2020.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/17/2020] [Accepted: 12/16/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND AIMS Distraction osteogenesis (DO) is a surgical technique to promote bone regeneration that requires a long time for bone healing. Bone marrow-derived mesenchymal stromal cells (MSCs) have been applied to accelerate bone formation in DO. Allogeneic MSCs are attractive, as they could be ready to use in clinics. Whether allogeneic MSCs would have an effect similar to autologous MSCs with regard to promoting bone formation in DO is still unknown. This study compares the effect of autologous MSCs versus allogeneic MSCs on bone formation in a rat DO model. METHODS Rat bone marrow-derived MSCs were isolated, characterized and expanded in vitro. Adult rats were subjected to right tibia transverse osteotomy. On the third day of distraction, each rat received one injection of phosphate-buffered saline (PBS), autologous MSCs or allogeneic MSCs at the distraction site. Tibiae were harvested after 28 days of consolidation for micro-computed tomography examination, mechanical test and histological analysis. RESULTS Results showed that treatment with both allogeneic and autologous MSCs promoted bone formation, with significantly higher bone mass, mechanical properties and mineral apposition rate as well as expression of angiogenic and bone formation markers at the regeneration sites compared with the PBS-treated group. No statistical difference in bone formation was found between the allogeneic and autologous MSC treatment groups. CONCLUSIONS This study indicates that allogeneic and autologous MSCs have a similar effect on promoting bone consolidation in DO. MSCs from an allogeneic source could be used off-the-shelf with DO to achieve early bone healing.
Collapse
Affiliation(s)
- Qi Pan
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Ye Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Yucong Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Haixing Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Lingchi Kong
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Zhengmeng Yang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Xiaoting Zhang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Shanshan Bai
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China
| | - Zhixian Zong
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Guanghua Chen
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Sien Lin
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China; Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, California, USA.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China; The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China; Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China; Department of Orthopaedics and Traumatology, Affiliated Baoan Hospital of Shenzhen, Southern Medical University, People's Hospital of Baoan District, Shenzhen, China..
| |
Collapse
|
7
|
Li Y, Pan Q, Xu J, He X, Li HA, Oldridge DA, Li G, Qin L. Overview of methods for enhancing bone regeneration in distraction osteogenesis: Potential roles of biometals. J Orthop Translat 2021; 27:110-118. [PMID: 33575164 PMCID: PMC7859169 DOI: 10.1016/j.jot.2020.11.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Distraction osteogenesis (DO) is a functional tissue engineering approach that applies gradual mechanical traction on the bone tissues after osteotomy to stimulate bone regeneration. However, DO still has disadvantages that limit its clinical use, including long treatment duration. METHODS Review the current methods of promoting bone formation and consolidation in DO with particular interest on biometal. RESULTS Numerous approaches, including physical therapy, gene therapy, growth factor-based therapy, stem-cell-based therapy, and improved distraction devices, have been explored to reduce the DO treatment duration with some success. Nevertheless, no approach to date is widely accepted in clinical practice due to various reasons, such as high expense, short biologic half-life, and lack of effective delivery methods. Biometals, including calcium (Ca), magnesium (Mg), zinc (Zn), copper (Cu), manganese (Mn), and cobalt (Co) have attracted attention in bone regeneration attributed to their biodegradability and bioactive components released during in vivo degradation. CONCLUSION This review summarizes the current therapies accelerating bone formation in DO and the beneficial role of biometals in bone regeneration, particularly focusing on the use of biometal Mg and its alloy in promoting bone formation in DO. Translational potential: The potential clinical applications using Mg-based devices to accelerate DO are promising. Mg stimulates expression of multiple intrinsic biological factors and the development of Mg as an implantable component in DO may be used to argument bone formation and consolidation in DO.
Collapse
Affiliation(s)
- Ye Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Qi Pan
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Xuan He
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Helen A. Li
- School of Medicine, University of East Anglia, Norwich, England, UK
| | - Derek A. Oldridge
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
8
|
Du GQ, Gong ZH, Liang B, Li P, Yang SY, Jia L, Jiang JH, Zhang K. Concentration Changes of Peripheral Blood Mesenchymal Stem Cells of Sprague Dawley Rats during Distraction Osteogenesis. Orthop Surg 2021; 13:623-631. [PMID: 33565272 PMCID: PMC7957402 DOI: 10.1111/os.12823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 09/06/2020] [Accepted: 09/16/2020] [Indexed: 01/26/2023] Open
Abstract
Objectives To observe the changes in the concentrations of circulating peripheral blood mesenchymal stem cells (PBMSCs) in Sprague Dawley (SD) rats and explore the pattern of changes in PBMSCs during the process of distraction osteogenesis. Methods SD rats were randomly divided into the osteotomy with lengthening group (lengthening group), the osteotomy without lengthening group (osteotomy group), and the blank control group (control group). Each group included 24 rats. Percutaneous pinning with external fixation of the left femur was carried out in lengthening group and osteotomy group, but control group received no surgical treatment. On day 5 after operation, continuous traction was carried out at a rate of 0.25 mm/d in lengthening group, while no traction was carried out in osteotomy group. Peripheral blood was collected from all rats on days 1, 3, 7, and 16 after the start of traction. PBMSCs were isolated by density gradient centrifugation. CD105, CD34, and CD45 were selected as cell surface markers. The concentration of PBMSCs was detected by flow cytometry and compared between groups at different time points. X‐ray films were taken during and after the operation to observe whether the osteotomy end was pulled and the growth and mineralization of the new bone in the osteogenic area of the femur. Color ultrasound was used to monitor the width of the distraction space, the formation of new bone, and the blood supply of soft tissue around the distraction. Results All rats were able to tolerate the operation well, and the external fixation was firm and reliable. X‐ray showed that, in lengthening group, the distraction space of femur gradually widened and new bone gradually formed in the distraction space; after 8 weeks, the samples were taken out, which showed that the new bone tissue in the lengthened area healed well. In osteotomy group, the average healing time of osteotomy was (7.12 ± 0.78) weeks. Ultrasonic examination showed that after the end of traction, the high echo callus shadow was seen in the traction space, and the blood flow signal was obviously rich at an earlier stage. In lengthening group and osteotomy group, the average concentrations of PBMSCs (3.02% ± 0.87% vs 2.95% ± 0.74%, respectively) were significantly increased in the early stage after osteotomy, and the average concentrations of PBMSCs on days 3, 7, and 16 after the start of traction were 5.34% ± 1.13% vs 3.28% ± 1.22%; 6.41% ± 1.05% vs 3.16% ± 0.92%; and 5.94% ± 1.23% vs 1.48% ± 0.52%, respectively. The concentration of PBMSCs in peripheral blood of lengthening group and osteotomy group was the same at osteotomy stage, and the difference between the two groups was not statistically significant (P > 0.05). After that, compared with lengthening group, the concentration of PBMSCs in osteotomy group gradually decreased and maintained at a certain level; the difference between the two groups was statistically significant (P < 0.05). Conclusions Distraction osteogenesis of femur can significantly increase PBMSCs in SD rats and participate in the process of bone formation.
Collapse
Affiliation(s)
- Gang-Qiang Du
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China
| | - Zhi-Hao Gong
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China
| | - Bin Liang
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China.,Department of Orthopaedic Surgery, The People's Hospital of Zhao Yuan City, Yantai, China
| | - Peng Li
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China
| | - Shu-Ye Yang
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China
| | - Long Jia
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China
| | - Jian-Hao Jiang
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China
| | - Kai Zhang
- Department of Orthopaedic Trauma, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
9
|
Yang Z, Xiao L, Deng Z, Cai L, Xie Y. Evaluation of Demineralized Bone Matrix Particles Delivered by Alginate Hydrogel for a Bone Graft Substitute: An Animal Experimental Study. Med Sci Monit 2021; 27:e928617. [PMID: 33481770 PMCID: PMC7836326 DOI: 10.12659/msm.928617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Our objective was to explore a synthetic alginate hydrogel delivery system for the delivery of demineralized bone matrix (DBM) particles for bone graft substitutes. MATERIAL AND METHODS The physiochemical properties of surface morphology, porosity measurements, in vitro degradation, equilibrium swelling, and mechanical testing of combined DBM powder and alginate in amounts of 0 mg/1 mL, 25 mg/1 mL, 50 mg/1 mL, and 100 mg/1 mL were detected. In vitro cell culture and in vivo studies using Sprague-Dawley rats were performed to evaluate the biocompatibility and osteoinductivity of DBM-alginate (ADBM) composites. RESULTS DBM particles were uniformly scattered in all composites, and macro-scale pores were omnipresent. All composites showed a similar low degradation rate, with approximately 85% of weight remaining after 15 days. As the concentration of DBM particles in composites increased, degradation in collagenase and elastic modulus increased and the pore area and swelling ratio significantly decreased. No cytotoxicity of ADBM or alginate on mesenchymal stem cells (MSCs) was observed. Cell cultivation with ADBM showed greater osteogenic potential, evidenced by the upregulation of alkaline phosphatase and alizarin red staining activity and the mRNA expression level of marker genes RUNX2, OCN, OPN, and collagen I compared with the cells grown in alginate. Evaluation of ectopic bone formation revealed the osteoinductivity of the ADBM composites was significantly greater than that of DBM particles. Osteoinduction of the composites was demonstrated by a cranial defect model study. CONCLUSIONS The delivery of DBM particles using a synthetic alginate hydrogel carrier may be a promising approach in bone tissue engineering for bone defects.
Collapse
Affiliation(s)
- Zhiqiang Yang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Yuanlong Xie
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
10
|
Yang Y, Pan Q, Zou K, Wang H, Zhang X, Yang Z, Lee WYW, Wei B, Gu W, Yang YP, Lin S, Li G. Administration of allogeneic mesenchymal stem cells in lengthening phase accelerates early bone consolidation in rat distraction osteogenesis model. Stem Cell Res Ther 2020; 11:129. [PMID: 32197646 PMCID: PMC7083044 DOI: 10.1186/s13287-020-01635-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/20/2020] [Accepted: 03/04/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Distraction osteogenesis (DO) is a surgical technique to promote bone regeneration which may require long duration for bone consolidation. Bone marrow-derived mesenchymal stem cells (MSCs) have been applied to accelerate bone formation in DO. However, the optimal time point for cell therapy in DO remains unknown. This study sought to determine the optimal time point of cell administration to achieve early bone consolidation in DO. We hypothesized that the ratio of circulating MSCs to peripheral mononuclear cells and the level of cytokines in serum might be indicators for cell administration in DO. METHODS Unilateral tibial osteotomy with an external fixator was performed in adult Sprague Dawley rats. Three days after osteotomy, the tibia was lengthened at 0.5 mm/12 h for 5 days. At first, 5 rats were used to analyze the blood components at 6 different time points (3 days before lengthening, on the day lengthening began, or 3, 6, 10, or 14 days after lengthening began) by sorting circulating MSCs and measuring serum levels of stromal cell-derived factor 1 (SDF-1) and interleukin 1β. Then, 40 rats were used for cell therapy study. A single dose of 5 × 105 allogeneic MSCs was locally injected at the lengthening site on day 3, 6, or 10 after lengthening began, or 3 doses of MSCs were injected at the three time points. Sequential X-ray radiographs were taken weekly. Endpoint examinations included micro-computed tomography analysis, mechanical testing, histomorphometry, and histology. RESULTS The number of circulating MSCs and serum level of SDF-1 were significantly increased during lengthening, and then decreased afterwards. Single injection of MSCs during lengthening phase (on day 3, but not day 6 or 10) significantly increased bone volume fraction, mechanical maximum loading, and bone mineralization of the regenerate. Triple injections of MSCs at three time points also significantly increased bone volume and maximum loading of the regenerates. CONCLUSION This study demonstrated that bone consolidation could be accelerated by a single injection of MSCs during lengthening when the ratio of peripheral MSCs to mononuclear cells and the serum SDF-1 presented at peak levels concurrently, suggesting that day 3 after lengthening began may be the optimal time point for cell therapy to promote early bone consolidation.
Collapse
Affiliation(s)
- Yanhua Yang
- Department of Central Laboratory, Changzhou Seventh People’s Hospital, Changzhou, China
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Qi Pan
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Kaijie Zou
- Department of Central Laboratory, Changzhou Seventh People’s Hospital, Changzhou, China
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Haixing Wang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Xiaoting Zhang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Zhengmeng Yang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Wayne Yuk Wai Lee
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Bo Wei
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Weidong Gu
- Department of Central Laboratory, Changzhou Seventh People’s Hospital, Changzhou, China
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, USA
- Department of Materials Science and Engineering, School of Engineering, Stanford University, Stanford, USA
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, USA
| | - Sien Lin
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, USA
| | - Gang Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
11
|
Medhat D, Rodríguez CI, Infante A. Immunomodulatory Effects of MSCs in Bone Healing. Int J Mol Sci 2019; 20:ijms20215467. [PMID: 31684035 PMCID: PMC6862454 DOI: 10.3390/ijms20215467] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into multilineage cells, thus making them a significant prospect as a cell source for regenerative therapy; however, the differentiation capacity of MSCs into osteoblasts seems to not be the main mechanism responsible for the benefits associated with human mesenchymal stem cells hMSCs when used in cell therapy approaches. The process of bone fracture restoration starts with an instant inflammatory reaction, as the innate immune system responds with cytokines that enhance and activate many cell types, including MSCs, at the site of the injury. In this review, we address the influence of MSCs on the immune system in fracture repair and osteogenesis. This paradigm offers a means of distinguishing target bone diseases to be treated with MSC therapy to enhance bone repair by targeting the crosstalk between MSCs and the immune system.
Collapse
Affiliation(s)
- Dalia Medhat
- Medical Biochemistry Department, National Research Centre, Dokki, Giza 12622, Egypt.
| | - Clara I Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| |
Collapse
|
12
|
The Effect of Bone Marrow Aspirate Concentrate Application on Distracted Bone Biomechanical Properties. J Craniofac Surg 2019; 30:2650-2655. [DOI: 10.1097/scs.0000000000005998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
13
|
Torres-Torrillas M, Rubio M, Damia E, Cuervo B, Del Romero A, Peláez P, Chicharro D, Miguel L, Sopena JJ. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of Musculoskeletal Diseases. Int J Mol Sci 2019; 20:ijms20123105. [PMID: 31242644 PMCID: PMC6627452 DOI: 10.3390/ijms20123105] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023] Open
Abstract
Chronic musculoskeletal (MSK) pain is one of the most common medical complaints worldwide and musculoskeletal injuries have an enormous social and economical impact. Current pharmacological and surgical treatments aim to relief pain and restore function; however, unsatiscactory outcomes are commonly reported. In order to find an accurate treatment to such pathologies, over the last years, there has been a significantly increasing interest in cellular therapies, such as adipose-derived mesenchymal stem cells (AMSCs). These cells represent a relatively new strategy in regenerative medicine, with many potential applications, especially regarding MSK disorders, and preclinical and clinical studies have demonstrated their efficacy in muscle, tendon, bone and cartilage regeneration. Nevertheless, several worries about their safety and side effects at long-term remain unsolved. This article aims to review the current state of AMSCs therapy in the treatment of several MSK diseases and their clinical applications in veterinary and human medicine.
Collapse
Affiliation(s)
- Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Monica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Elena Damia
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Belen Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Ayla Del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Laura Miguel
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Joaquin J Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| |
Collapse
|
14
|
Montes-Medina L, Hernández-Fernández A, Gutiérrez-Rivera A, Ripalda-Cemboráin P, Bitarte N, Pérez-López V, Granero-Moltó F, Prosper F, Izeta A. Effect of bone marrow stromal cells in combination with biomaterials in early phases of distraction osteogenesis: An experimental study in a rabbit femur model. Injury 2018; 49:1979-1986. [PMID: 30219381 DOI: 10.1016/j.injury.2018.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 08/24/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023]
Abstract
Acceleration of the consolidation of the distracted bone is a relevant medical need. As a platform to improve in vivo bone engineering, we developed a novel distraction osteogenesis (DO) model in a rabbit large bone (femur) and tested if the application of cultured bone marrow stromal cells (BMSCs) immediately after the osteotomy promotes the formation of bone. This report consists of two components, an animal study to evaluate the quality of the regenerate following different treatments and an in vitro study to evaluate osteogenic potential of BMSC cultures. To illuminate the mechanism of action of injected cells, we tested stem cell cultures enriched in osteogenic-BMSCs (O-BMSCs) as compared with cultures enriched in non-osteogenic BMSCs (NO-BMSCs). Finally, we included a group of animals treated with biomaterials (fibrin and ground cortical bone) in addition to cells. Injection of O-BMSCs promoted the maturity of distracted callus and decreased fibrosis. When combined with biomaterials, O-BMSCs modified the ossification pattern from endochondral to intramembranous type. The use of NO-BMSCs not only did not increase the maturity but also increased porosity of the bone. These preclinical results indicate that the BMSC cultures must be tested in vitro prior to clinical use, since a number of factors may influence their outcome in bone formation. We hypothesize that the use of osteogenic BMSCs and biomaterials could be clinically beneficial to shorten the consolidation period of the distraction and the total period of bone lengthening.
Collapse
Affiliation(s)
- Laura Montes-Medina
- Department of Orthopaedic Surgery, Donostia University Hospital, San Sebastian, Spain
| | - Alberto Hernández-Fernández
- Department of Orthopaedic Surgery, Donostia University Hospital, San Sebastian, Spain; Department of Surgery, Radiology and Physical Medicine of the University of the Basque Country (UPV-EHU), San Sebastian, Spain
| | | | | | - Nerea Bitarte
- Tissue Engineering Group, Bioengineering Area, Instituto Biodonostia, San Sebastian, Spain
| | - Virginia Pérez-López
- Tissue Engineering Group, Bioengineering Area, Instituto Biodonostia, San Sebastian, Spain
| | - Froilán Granero-Moltó
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain; Cell Therapy Area, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Cell Therapy Area, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Ander Izeta
- Tissue Engineering Group, Bioengineering Area, Instituto Biodonostia, San Sebastian, Spain; Department of Biomedical Engineering and Science, School of Engineering, Tecnun-University of Navarra, San Sebastian, Spain.
| |
Collapse
|
15
|
Zomer HD, Roballo KC, Lessa TB, Bressan FF, Gonçalves NN, Meirelles FV, Trentin AG, Ambrósio CE. Distinct features of rabbit and human adipose-derived mesenchymal stem cells: implications for biotechnology and translational research. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:43-54. [PMID: 30425533 PMCID: PMC6204872 DOI: 10.2147/sccaa.s175749] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction Owing to their similarity with humans, rabbits are useful for multiple applications in biotechnology and translational research from basic to preclinical studies. In this sense, mesenchymal stem cells (MSCs) are known for their therapeutic potential and promising future in regenerative medicine. As many studies have been using rabbit adipose-derived MSCs (ASCs) as a model of human ASCs (hASCs), it is fundamental to compare their characteristics and understand how distinct features could affect the translation to human medicine. Objective The aim of this study was to comparatively characterize rabbit ASCs (rASCs) and hASCs to further uses in biotechnology and translational studies. Materials and methods rASCs and hASCs were isolated and characterized by their immunophenotype, differentiation potential, proliferative profile, and nuclear stability in vitro. Results and discussion Both ASCs presented differentiation potential to osteocytes, chondrocytes, and adipocytes and shared similar immunophenotype expression to CD105+, CD34−, and CD45−, but rabbit cells expressed significantly lower CD73 and CD90 than human cells. In addition, rASCs presented greater clonogenic potential and proliferation rate than hASCs but no difference in nuclear alterations. Conclusion The distinct features of rASCs and hASCs can positively or negatively affect their use for different applications in biotechnology (such as cell reprogramming) and translational studies (such as cell transplantation, tissue engineering, and pharmacokinetics). Nevertheless, the particularities between rabbit and human MSCs should not prevent rabbit use in preclinical models, but care should be taken to interpret results and properly translate animal findings to medicine.
Collapse
Affiliation(s)
- Helena Debiazi Zomer
- Department of Cell Biology, Embryology and Genetic, Faculty of Biological Sciences, Santa Catarina Federal University (UFSC), Florianópolis, Brazil.,Department of Surgery, Sector Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil,
| | - Kelly Cs Roballo
- Department of Surgery, Sector Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil,
| | - Thais Borges Lessa
- Department of Surgery, Sector Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil,
| | - Fabiana Fernandes Bressan
- Department of Surgery, Sector Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil,
| | - Natália Nardeli Gonçalves
- Department of Surgery, Sector Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil,
| | - Flávio Vieira Meirelles
- Department of Surgery, Sector Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil, .,Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil,
| | - Andrea Gonçalves Trentin
- Department of Cell Biology, Embryology and Genetic, Faculty of Biological Sciences, Santa Catarina Federal University (UFSC), Florianópolis, Brazil
| | - Carlos Eduardo Ambrósio
- Department of Surgery, Sector Anatomy, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil, .,Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil,
| |
Collapse
|
16
|
Jin YZ, Lee JH. Mesenchymal Stem Cell Therapy for Bone Regeneration. Clin Orthop Surg 2018; 10:271-278. [PMID: 30174801 PMCID: PMC6107811 DOI: 10.4055/cios.2018.10.3.271] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/27/2018] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in clinic for approximately 20 years. During this period, various new populations of MSCs have been found or manipulated. However, their characters and relative strength for bone regeneration have not been well known. For a comprehensive understanding of MSCs, we reviewed the literature on the multipotent cells ranging from the definition to the current research progress for bone regeneration. Based on our literature review, bone marrow MSCs have been most widely studied and utilized in clinical settings. Among other populations of MSCs, adipose-derived MSCs and perivascular MSCs might be potential candidates for bone regeneration, whose efficacy and safety still require further investigation.
Collapse
Affiliation(s)
- Yuan-Zhe Jin
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Hyup Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea.,Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul, Korea.,Institute of Medical and Biological Engineering, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
17
|
Amuk NG, Kurt G, Baran Y, Seyrantepe V, Yandim MK, Adan A, Demir SA, Kiraz Y, Sonmez MF. Effects of cell-mediated osteoprotegerin gene transfer and mesenchymal stem cell applications on orthodontically induced root resorption of rat teeth. Eur J Orthod 2018; 39:235-242. [PMID: 27733487 DOI: 10.1093/ejo/cjw054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aim The aim of this study is to evaluate and compare therapeutic effects of mesenchymal stem cell (MSCs) and osteoprotegerin (OPG) gene transfer applications on inhibition and/or repair of orthodontically induced inflammatory root resorption (OIIRR). Materials and methods Thirty Wistar rats were divided into four groups as untreated group (negative control), treated with orthodontic appliance group (positive control), MSCs injection group, and OPG transfected MSCs [gene therapy (GT) group]. About 100g of orthodontic force was applied to upper first molar teeth of rats for 14 days. MSCs and transfected MSC injections were performed at 1st, 6th, and 11th days to the MSC and GT group rats. At the end of experiment, upper first molar teeth were prepared for genetical, scanning electron microscopy (SEM), fluorescent microscopy, and haematoxylin eosin-tartrate resistant acid phosphatase staining histological analyses. Number of total cells, number of osteoclastic cells, number of resorption lacunae, resorption area ratio, SEM resorption ratio, OPG, RANKL, Cox-2 gene expression levels at the periodontal ligament (PDL) were calculated. Paired t-test, Kruskal-Wallis, and chi-square tests were performed. Results Transferred MSCs showed marked fluorescence in PDL. The results revealed that number of osteoclastic cells, resorption lacunae, resorption area ratio, RANKL, and Cox-2 were reduced after single MSC injections significantly (P < 0.05). GT group showed the lowest number of osteoclastic cells (P < 0.01), number of resorption lacunae, resorption area ratio, and highest OPG expression (P < 0.001). Conclusions Taken together all these results, MSCs and GT showed marked inhibition and/or repair effects on OIIRR during orthodontic treatment on rats.
Collapse
Affiliation(s)
- Nisa Gul Amuk
- Department of Orthodontics, Faculty of Dentistry, Erciyes University, Kayseri
| | - Gökmen Kurt
- Department of Orthodontics, Faculty of Dentistry, Istanbul Yeni Yüzyil University
| | - Yusuf Baran
- Department of Molecular Biology and Genetics, Faculty of Science, Izmir Institute of Technology.,Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri
| | - Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Faculty of Science, Izmir Institute of Technology
| | - Melis Kartal Yandim
- Department of Molecular Biology and Genetics, Faculty of Science, Izmir Institute of Technology.,Department of Medical biology, Faculty of Medicine, Izmir University of Economics
| | - Aysun Adan
- Department of Molecular Biology and Genetics, Faculty of Science, Izmir Institute of Technology.,Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri
| | - Secil Akyildiz Demir
- Department of Molecular Biology and Genetics, Faculty of Science, Izmir Institute of Technology
| | - Yagmur Kiraz
- Department of Molecular Biology and Genetics, Faculty of Science, Izmir Institute of Technology.,Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri
| | - Mehmet Fatih Sonmez
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
18
|
Safari S, Mahdian A, Motamedian SR. Applications of stem cells in orthodontics and dentofacial orthopedics: Current trends and future perspectives. World J Stem Cells 2018; 10:66-77. [PMID: 29988866 PMCID: PMC6033713 DOI: 10.4252/wjsc.v10.i6.66] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/19/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023] Open
Abstract
A simple overview of daily orthodontic practice involves use of brackets, wires and elastomeric modules. However, investigating the underlying effect of orthodontic forces shows various molecular and cellular changes. Also, orthodontics is in close relation with dentofacial orthopedics which involves bone regeneration. In this review current and future applications of stem cells (SCs) in orthodontics and dentofacial orthopedics have been discussed. For craniofacial anomalies, SCs have been applied to regenerate hard tissue (such as treatment of alveolar cleft) and soft tissue (such as treatment of hemifacial macrosomia). Several attempts have been done to reconstruct impaired temporomandibular joint. Also, SCs with or without bone scaffolds and growth factors have been used to regenerate bone following distraction osteogenesis of mandibular bone or maxillary expansion. Current evidence shows that SCs also have potential to be used to regenerate infrabony alveolar defects and move the teeth into regenerated areas. Future application of SCs in orthodontics could involve accelerating tooth movement, regenerating resorbed roots and expanding tooth movement limitations. However, evidence supporting these roles is weak and further studies are required to evaluate the possibility of these ideas.
Collapse
Affiliation(s)
- Shiva Safari
- Department of Orthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran 13819, Iran
| | - Arezoo Mahdian
- Department of Orthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran 13819, Iran
| | - Saeed Reza Motamedian
- Department of Orthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran 13819, Iran.
| |
Collapse
|
19
|
Yang Y, Lin S, Wang B, Gu W, Li G. Stem cell therapy for enhancement of bone consolidation in distraction osteogenesis: A contemporary review of experimental studies. Bone Joint Res 2017; 6:385-390. [PMID: 28634158 PMCID: PMC5492338 DOI: 10.1302/2046-3758.66.bjr-2017-0023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/11/2017] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES Distraction osteogenesis (DO) mobilises bone regenerative potential and avoids the complications of other treatments such as bone graft. The major disadvantage of DO is the length of time required for bone consolidation. Mesenchymal stem cells (MSCs) have been used to promote bone formation with some good results. METHODS We hereby review the published literature on the use of MSCs in promoting bone consolidation during DO. RESULTS Studies differed in animal type (mice, rabbit, dog, sheep), bone type (femur, tibia, skull), DO protocols and cell transplantation methods. CONCLUSION The majority of studies reported that the transplantation of MSCs enhanced bone consolidation or formation in DO. Many questions relating to animal model, DO protocol and cell transplantation regime remain to be further investigated. Clinical trials are needed to test and confirm these findings from animal studies.Cite this article: Y. Yang, S. Lin, B. Wang, W. Gu, G. Li. Stem cell therapy for enhancement of bone consolidation in distraction osteogenesis: A contemporary review of experimental studies. Bone Joint Res 2017;6:385-390. DOI: 10.1302/2046-3758.66.BJR-2017-0023.
Collapse
Affiliation(s)
- Y Yang
- Department of Key Laboratory, Changzhou No.7 People's Hospital, No. 288 Yanling East Road, Changzhou, Jiangsu, China
| | - S Lin
- Department of Orthopaedics and Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - B Wang
- Department of Orthopaedics and Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - W Gu
- Department of Traumatology, Changzhou No.7 People's Hospital, No. 288 Yanling East Road, Changzhou, Jiangsu, China
| | - G Li
- Department of Orthopaedics and Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| |
Collapse
|
20
|
Barba M, Di Taranto G, Lattanzi W. Adipose-derived stem cell therapies for bone regeneration. Expert Opin Biol Ther 2017; 17:677-689. [PMID: 28374644 DOI: 10.1080/14712598.2017.1315403] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell-based therapies exploit the heterogeneous and self-sufficient biological environment of stem cells to restore, maintain and improve tissue functions. Adipose-derived stem cells (ASCs) are, to this aim, promising cell types thanks to advantageous isolation procedures, growth kinetics, plasticity and trophic properties. Specifically, bone regeneration represents a suitable, though often challenging, target setting to test and apply ASC-based therapeutic strategies. Areas covered: ASCs are extremely plastic and secrete bioactive peptides that mediate paracrine functions, mediating their trophic actions in vivo. Numerous preclinical studies demonstrated that ASCs improve bone healing. Clinical trials are ongoing to validate the clinical feasibility of these approaches. This review is intended to define the state-of-the-art on ASCs, encompassing the biological features that make them suitable for bone regenerative strategies, and to provide an update on existing preclinical and clinical applications. Expert opinion: ASCs offer numerous advantages over other stem cells in terms of feasibility of clinical translation. Data obtained from in vivo experimentation are encouraging, and clinical trials are ongoing. More robust validations are thus expected to be achieved during the next few years, and will likely pave the way to optimized patient-tailored treatments for bone regeneration.
Collapse
Affiliation(s)
- Marta Barba
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giuseppe Di Taranto
- b Department of Plastic, Reconstructive and Aesthetic Surgery , University of Rome "Sapienza" , Policlinico Umberto I, Rome , Italy
| | - Wanda Lattanzi
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
21
|
Guerado E, Caso E. Challenges of bone tissue engineering in orthopaedic patients. World J Orthop 2017; 8:87-98. [PMID: 28251059 PMCID: PMC5314152 DOI: 10.5312/wjo.v8.i2.87] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/23/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
Bone defects may impede normal biomechanics and the structural stability of bone as an organ. In many cases, the correction of bone defects requires extensive surgical intervention involving the use of bone-grafting techniques and other procedures in which healing is slow, there is a high risk of infection and considerable pain is provoked - with no guarantee of complete correction of the defect. Therefore, the search for surgical alternatives continues to present a major challenge in orthopaedic traumatology. The reamer-irrigator-aspirator (RIA) system, which was devised to avoid the problems that can arise with autograft harvesting from the iliac crest, consists of collecting the product of the femoral canal after reaming. The RIA technique improves osteogenic differentiation of mesenchymal stem cells, compared to bone marrow aspiration or cancellous bone harvesting from the iliac crest using a spoon. Another approach, the Masquelet technique, consists of reconstructing a long bone defect by means of an induced membrane grown onto an acrylic cement rod inserted to fill the defect; in a second surgical step, once the membrane is constituted, the cement rod is removed and cancellous autograft is used to fill the defect. Both in RIA and in the Masquelet technique, osteosynthesis is usually needed. Bone transportation by compression-distraction lengthening principles is commonly implemented for the treatment of large bone loss. However, complications are frequently encountered with these techniques. Among new techniques that have been proposed to address the problem of large bone loss, the application of stem cells in conjunction with tissue engineering techniques is very promising, as is the creation of personalised medicine (or precision medicine), in which molecular profiling technologies are used to tailor the therapeutic strategy, to ensure the right method is applied for the right person at the right time, after determining the predisposition to disease among the general population. All of the above techniques for addressing bone defects are discussed in this paper.
Collapse
|
22
|
Abstract
Unlike many other postnatal tissues, bone can regenerate and repair itself; nevertheless, this capacity can be overcome. Traditionally, surgical reconstructive strategies have implemented autologous, allogeneic, and prosthetic materials. Autologous bone--the best option--is limited in supply and also mandates an additional surgical procedure. In regenerative tissue engineering, there are myriad issues to consider in the creation of a functional, implantable replacement tissue. Importantly, there must exist an easily accessible, abundant cell source with the capacity to express the phenotype of the desired tissue, and a biocompatible scaffold to deliver the cells to the damaged region. A literature review was performed using PubMed; peer-reviewed publications were screened for relevance in order to identify key advances in stem and progenitor cell contribution to the field of bone tissue engineering. In this review, we briefly introduce various adult stem cells implemented in bone tissue engineering such as mesenchymal stem cells (including bone marrow- and adipose-derived stem cells), endothelial progenitor cells, and induced pluripotent stem cells. We then discuss numerous advances associated with their application and subsequently focus on technological advances in the field, before addressing key regenerative strategies currently used in clinical practice. Stem and progenitor cell implementation in bone tissue engineering strategies have the ability to make a major impact on regenerative medicine and reduce patient morbidity. As the field of regenerative medicine endeavors to harness the body's own cells for treatment, scientific innovation has led to great advances in stem cell-based therapies in the past decade.
Collapse
|
23
|
Regeneration of mandibular defects using adipose tissue mesenchymal stromal cells in combination with human serum-derived scaffolds. J Craniomaxillofac Surg 2016; 44:1356-65. [PMID: 27450897 DOI: 10.1016/j.jcms.2016.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 05/17/2016] [Accepted: 06/13/2016] [Indexed: 12/19/2022] Open
|
24
|
Işık S, Karaman M, Adan A, Kıray M, Bağrıyanık HA, Sözmen ŞÇ, Kozanoğlu İ, Karaman Ö, Baran Y, Uzuner N. Intraperitoneal mesenchymal stem cell administration ameliorates allergic rhinitis in the murine model. Eur Arch Otorhinolaryngol 2016; 274:197-207. [PMID: 27380271 DOI: 10.1007/s00405-016-4166-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/22/2016] [Indexed: 12/26/2022]
Abstract
Previous studies showed that bone marrow-derived mesenchymal stem cells (BMSCs) could ameliorate a variety of immune-mediated and inflammatory diseases due to their immunomodulatory and anti-inflammatory effects. In this study, we developed a mouse model of ovalbumin (OVA) induced allergic inflammation in the upper airways and evaluated the effects of the intraperitoneal administration of BMSCs on allergic inflammation. Twenty-five BALB/c mice were divided into five groups; group I (control group), group II (sensitized and challenged with OVA and treated with saline-placebo group), group III (sensitized and challenged with OVA and treated with 1 × 106 BMSCs), group IV (sensitized and challenged with OVA and treated with 2 × 106 BMSCs), and group V (sensitized and challenged with phosphate buffered saline (PBS) and treated with 1 × 106 BMSCs). Histopathological features (number of goblet cells, eosinophils and mast cells, basement membrane, epithelium thickness, and subepithelial smooth muscle thickness) of the upper and lower airways and BMSCs migration to nasal and lung tissue were evaluated using light and confocal microscopes. Levels of cytokines in the nasal lavage fluid and lung tissue supernatants were measured using enzyme-linked immunosorbent assay (ELISA). Confocal microscopic analysis showed that there was no significant amount of BMSCs in the nasal and lung tissues of group V. However, significant amount of BMSCs were observed in group III and IV. In OVA-induced AR groups (group II, III, and IV), histopathological findings of chronic asthma, such as elevated subepithelial smooth muscle thickness, epithelium thickness, and number of goblet and mast cells, were determined. Furthermore, the number of nasal goblet and eosinophil cells, histopathological findings of chronic asthma, and IL-4, IL-5, IL-13, and NO levels was significantly lower in both BMSCs-treated groups compared to the placebo group. Our findings indicated that histopathological findings of chronic asthma were also observed in mice upon AR induction. BMSCs migrated to the nasal and lung tissues following intraperitoneal delivery and ameliorated to the airway remodeling and airway inflammation both in the upper and lower airways via the inhibition of T helper (Th) 2 immune response in the murine model of AR.
Collapse
Affiliation(s)
- Sakine Işık
- Department of Pediatric Allergy and Immunology, Dokuz Eylul University, Balçova, 35330, Izmir, Turkey.
| | - Meral Karaman
- Department of Microbiology, Dokuz Eylul University, Izmir, Turkey
| | - Aysun Adan
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Müge Kıray
- Department of Physiology, Dokuz Eylul University, Izmir, Turkey
| | | | - Şule Çağlayan Sözmen
- Department of Pediatric Allergy and Immunology, Dokuz Eylul University, Balçova, 35330, Izmir, Turkey
| | | | - Özkan Karaman
- Department of Pediatric Allergy and Immunology, Dokuz Eylul University, Balçova, 35330, Izmir, Turkey
| | - Yusuf Baran
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Nevin Uzuner
- Department of Pediatric Allergy and Immunology, Dokuz Eylul University, Balçova, 35330, Izmir, Turkey
| |
Collapse
|
25
|
Vachkova E, Bosnakovski D, Yonkova P, Grigorova N, Ivanova Z, Todorov P, Penchev G, Milanova A, Simeonova G, Stanilova S, Georgiev IP. Adipogenic potential of stem cells derived from rabbit subcutaneous and visceral adipose tissue in vitro. In Vitro Cell Dev Biol Anim 2016; 52:829-37. [PMID: 27173612 DOI: 10.1007/s11626-016-0048-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/22/2016] [Indexed: 02/06/2023]
Abstract
Rabbits are considered as appropriate animal models to study some obesity-associated abnormalities because of the similarity of their blood lipid profile and metabolism to humans. The current study was focused on comparison of adipose differentiation ability in rabbit adipose-derived stem cells (ADSC) in vitro. Subcutaneous and visceral stromal vascular fractions (SVF) were isolated from three 28-d-old New Zealand rabbits by collagenase digestion. Supernatants from both isolates were collected 24 h after the initial plating. On the fourth passage, all isolated cell types undergo triplicate adipogenic induction. The adipose induction potential was calculated as percentage of increasing optical density (OD) values. The data revealed that with increasing the number of induction cycles, the induction tendency in visceral ADSC decreased in contrast to the subcutaneous ones. Although the supernatants did not reach induction levels of their relevant precursors, they follow the same pattern in both subcutaneous and visceral ADSC. All cell types successfully passed osteogenic and chondrogenic differentiation. In conclusion, the best adipose induction ability was observed in directly plated subcutaneous cell population. The increase of induction numbers depressed adipose induction ability in cell populations derived from visceral fat depots.
Collapse
Affiliation(s)
- Ekaterina Vachkova
- Animal Physiology Unit, Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6 000, Stara Zagora, Bulgaria.
| | - D Bosnakovski
- Faculty of Medical Sciences, University Goce Delčev-Štip, Shtip, Republic of Macedonia
| | - P Yonkova
- Department of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Trakia University, Stara Zagora, Bulgaria
| | - N Grigorova
- Animal Physiology Unit, Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6 000, Stara Zagora, Bulgaria
| | - Zh Ivanova
- Animal Physiology Unit, Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6 000, Stara Zagora, Bulgaria
| | - P Todorov
- Institute of Biology and Immunology of Reproduction, Sofia, Bulgaria
| | - G Penchev
- Department of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Trakia University, Stara Zagora, Bulgaria
| | - A Milanova
- Animal Physiology Unit, Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6 000, Stara Zagora, Bulgaria
| | - G Simeonova
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Trakia University, 6000, Stara Zagora, Bulgaria
| | - S Stanilova
- Department of Molecular Biology, Immunology and Genetics, Faculty of Medicine, Trakia University, Stara Zagora, Bulgaria
| | - I Penchev Georgiev
- Animal Physiology Unit, Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6 000, Stara Zagora, Bulgaria
| |
Collapse
|
26
|
Zomer HD, Vidane AS, Gonçalves NN, Ambrósio CE. Mesenchymal and induced pluripotent stem cells: general insights and clinical perspectives. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:125-34. [PMID: 26451119 PMCID: PMC4592031 DOI: 10.2147/sccaa.s88036] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells have awakened a great deal of interest in regenerative medicine due to their plasticity, and immunomodulatory and anti-inflammatory properties. They are high-yield and can be acquired through noninvasive methods from adult tissues. Moreover, they are nontumorigenic and are the most widely studied. On the other hand, induced pluripotent stem (iPS) cells can be derived directly from adult cells through gene reprogramming. The new iPS technology avoids the embryo destruction or manipulation to generate pluripotent cells, therefore, are exempt from ethical implication surrounding embryonic stem cell use. The pre-differentiation of iPS cells ensures the safety of future approaches. Both mesenchymal stem cells and iPS cells can be used for autologous cell transplantations without the risk of immune rejection and represent a great opportunity for future alternative therapies. In this review we discussed the therapeutic perspectives using mesenchymal and iPS cells.
Collapse
Affiliation(s)
- Helena D Zomer
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Atanásio S Vidane
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Natalia N Gonçalves
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Carlos E Ambrósio
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| |
Collapse
|
27
|
Comprehensive Review of Adipose Stem Cells and Their Implication in Distraction Osteogenesis and Bone Regeneration. BIOMED RESEARCH INTERNATIONAL 2015; 2015:842975. [PMID: 26448947 PMCID: PMC4584039 DOI: 10.1155/2015/842975] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/02/2015] [Indexed: 12/31/2022]
Abstract
Bone is one of the most dynamic tissues in the human body that can heal following injury without leaving a scar. However, in instances of extensive bone loss, this intrinsic capacity of bone to heal may not be sufficient and external intervention becomes necessary. Several techniques are available to address this problem, including autogenous bone grafts and allografts. However, all these techniques have their own limitations. An alternative method is the technique of distraction osteogenesis, where gradual and controlled distraction of two bony segments after osteotomy leads to induction of new bone formation. Although distraction osteogenesis usually gives satisfactory results, its major limitation is the prolonged duration of time required before the external fixator is removed, which may lead to numerous complications. Numerous methods to accelerate bone formation in the context of distraction osteogenesis have been reported. A viable alternative to autogenous bone grafts for a source of osteogenic cells is mesenchymal stem cells from bone marrow. However, there are certain problems with bone marrow aspirate. Hence, scientists have investigated other sources for mesenchymal stem cells, specifically adipose tissue, which has been shown to be an excellent source of mesenchymal stem cells. In this paper, the potential use of adipose stem cells to stimulate bone formation is discussed.
Collapse
|
28
|
Tevlin R, Atashroo D, Duscher D, Mc Ardle A, Gurtner GC, Wan DC, Longaker MT. Impact of surgical innovation on tissue repair in the surgical patient. Br J Surg 2015; 102:e41-55. [PMID: 25627135 DOI: 10.1002/bjs.9672] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/09/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Throughout history, surgeons have been prolific innovators, which is hardly surprising as most surgeons innovate daily, tailoring their intervention to the intrinsic uniqueness of each operation, each patient and each disease. Innovation can be defined as the application of better solutions that meet new requirements, unarticulated needs or existing market needs. In the past two decades, surgical innovation has significantly improved patient outcomes, complication rates and length of hospital stay. There is one key area that has great potential to change the face of surgical practice and which is still in its infancy: the realm of regenerative medicine and tissue engineering. METHODS A literature review was performed using PubMed; peer-reviewed publications were screened for relevance in order to identify key surgical innovations influencing regenerative medicine, with a focus on osseous, cutaneous and soft tissue reconstruction. RESULTS This review describes recent advances in regenerative medicine, documenting key innovations in osseous, cutaneous and soft tissue regeneration that have brought regenerative medicine to the forefront of the surgical imagination. CONCLUSION Surgical innovation in the emerging field of regenerative medicine has the ability to make a major impact on surgery on a daily basis.
Collapse
Affiliation(s)
- R Tevlin
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford, California, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Asatrian G, Pham D, Hardy WR, James AW, Peault B. Stem cell technology for bone regeneration: current status and potential applications. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:39-48. [PMID: 25709479 PMCID: PMC4334288 DOI: 10.2147/sccaa.s48423] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Continued improvements in the understanding and application of mesenchymal stem cells (MSC) have revolutionized tissue engineering. This is particularly true within the field of skeletal regenerative medicine. However, much remains unknown regarding the native origins of MSC, the relative advantages of different MSC populations for bone regeneration, and even the biologic safety of such unpurified, grossly characterized cells. This review will first summarize the initial discovery of MSC, as well as the current and future applications of MSC in bone tissue engineering. Next, the relative advantages and disadvantages of MSC isolated from distinct tissue origins are debated, including the MSC from adipose, bone marrow, and dental pulp, among others. The perivascular origin of MSC is next discussed. Finally, we briefly comment on pluripotent stem cell populations and their possible application in bone tissue engineering. While continually expanding, the field of MSC-based bone tissue engineering and regeneration shows potential to become a clinical reality in the not-so-distant future.
Collapse
Affiliation(s)
- Greg Asatrian
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, Los Angeles, CA, USA
| | - Dalton Pham
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, Los Angeles, CA, USA ; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Winters R Hardy
- UCLA/Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA, USA
| | - Aaron W James
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, Los Angeles, CA, USA ; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, Los Angeles, CA, USA ; UCLA/Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA, USA
| | - Bruno Peault
- UCLA/Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA, USA ; Medical Research Council Centre for Regenerative Medicine, Edinburgh, Scotland, UK
| |
Collapse
|
30
|
Wang X, Schröder HC, Grebenjuk V, Diehl-Seifert B, Mailänder V, Steffen R, Schloßmacher U, Müller WEG. The marine sponge-derived inorganic polymers, biosilica and polyphosphate, as morphogenetically active matrices/scaffolds for the differentiation of human multipotent stromal cells: potential application in 3D printing and distraction osteogenesis. Mar Drugs 2014; 12:1131-1147. [PMID: 24566262 PMCID: PMC3944534 DOI: 10.3390/md12021131] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/10/2014] [Accepted: 02/17/2014] [Indexed: 01/03/2023] Open
Abstract
The two marine inorganic polymers, biosilica (BS), enzymatically synthesized from ortho-silicate, and polyphosphate (polyP), a likewise enzymatically synthesized polymer consisting of 10 to >100 phosphate residues linked by high-energy phosphoanhydride bonds, have previously been shown to display a morphogenetic effect on osteoblasts. In the present study, the effect of these polymers on the differential differentiation of human multipotent stromal cells (hMSC), mesenchymal stem cells, that had been encapsulated into beads of the biocompatible plant polymer alginate, was studied. The differentiation of the hMSCs in the alginate beads was directed either to the osteogenic cell lineage by exposure to an osteogenic medium (mineralization activation cocktail; differentiation into osteoblasts) or to the chondrogenic cell lineage by incubating in chondrocyte differentiation medium (triggering chondrocyte maturation). Both biosilica and polyP, applied as Ca²⁺ salts, were found to induce an increased mineralization in osteogenic cells; these inorganic polymers display also morphogenetic potential. The effects were substantiated by gene expression studies, which revealed that biosilica and polyP strongly and significantly increase the expression of bone morphogenetic protein 2 (BMP-2) and alkaline phosphatase (ALP) in osteogenic cells, which was significantly more pronounced in osteogenic versus chondrogenic cells. A differential effect of the two polymers was seen on the expression of the two collagen types, I and II. While collagen Type I is highly expressed in osteogenic cells, but not in chondrogenic cells after exposure to biosilica or polyP, the upregulation of the steady-state level of collagen Type II transcripts in chondrogenic cells is comparably stronger than in osteogenic cells. It is concluded that the two polymers, biosilica and polyP, are morphogenetically active additives for the otherwise biologically inert alginate polymer. It is proposed that alginate, supplemented with polyP and/or biosilica, is a suitable biomaterial that promotes the growth and differentiation of hMSCs and might be beneficial for application in 3D tissue printing of hMSCs and for the delivery of hMSCs in fractures, surgically created during distraction osteogenesis.
Collapse
Affiliation(s)
- Xiaohong Wang
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Heinz C Schröder
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Vladislav Grebenjuk
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | | | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany.
| | - Renate Steffen
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Ute Schloßmacher
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Werner E G Müller
- ERC Advanced Investigator Grant Research Group, Institute for Physiological Chemistry, University Medical Center, Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| |
Collapse
|
31
|
Sun Z, Tee BC, Kennedy KS, Kennedy PM, Kim DG, Mallery SR, Fields HW. Scaffold-based delivery of autologous mesenchymal stem cells for mandibular distraction osteogenesis: preliminary studies in a porcine model. PLoS One 2013; 8:e74672. [PMID: 24040314 PMCID: PMC3764039 DOI: 10.1371/journal.pone.0074672] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/02/2013] [Indexed: 01/12/2023] Open
Abstract
Purpose Bone regeneration through distraction osteogenesis (DO) is promising but remarkably slow. To accelerate it, autologous mesenchymal stem cells have been directly injected to the distraction site in a few recent studies. Compared to direct injection, a scaffold-based method can provide earlier cell delivery with potentially better controlled cell distribution and retention. This pilot project investigated a scaffold-based cell-delivery approach in a porcine mandibular DO model. Materials and Methods Eleven adolescent domestic pigs were used for two major sets of studies. The in-vitro set established methodologies to: aspirate bone marrow from the tibia; isolate, characterize and expand bone marrow-derived mesenchymal stem cells (BM-MSCs); enhance BM-MSC osteogenic differentiation using FGF-2; and confirm cell integration with a gelatin-based Gelfoam scaffold. The in-vivo set transplanted autologous stem cells into the mandibular distraction sites using Gelfoam scaffolds; completed a standard DO-course and assessed bone regeneration by macroscopic, radiographic and histological methods. Repeated-measure ANOVAs and t-tests were used for statistical analyses. Results From aspirated bone marrow, multi-potent, heterogeneous BM-MSCs purified from hematopoietic stem cell contamination were obtained. FGF-2 significantly enhanced pig BM-MSC osteogenic differentiation and proliferation, with 5 ng/ml determined as the optimal dosage. Pig BM-MSCs integrated readily with Gelfoam and maintained viability and proliferative ability. After integration with Gelfoam scaffolds, 2.4–5.8×107 autologous BM-MSCs (undifferentiated or differentiated) were transplanted to each experimental DO site. Among 8 evaluable DO sites included in the final analyses, the experimental DO sites demonstrated less interfragmentary mobility, more advanced gap obliteration, higher mineral content and faster mineral apposition than the control sites, and all transplanted scaffolds were completely degraded. Conclusion It is technically feasible and biologically sound to deliver autologous BM-MSCs to the distraction site immediately after osteotomy using a Gelfoam scaffold to enhance mandibular DO.
Collapse
Affiliation(s)
- Zongyang Sun
- Division of Orthodontics, College of Dentistry, Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| | - Boon Ching Tee
- Division of Orthodontics, College of Dentistry, Ohio State University, Columbus, Ohio, United States of America
| | - Kelly S. Kennedy
- Division of Oral and Maxillofacial Surgery, College of Dentistry, Ohio State University, Columbus, Ohio, United States of America
| | - Patrick M. Kennedy
- Division of Oral and Maxillofacial Surgery, College of Dentistry, Ohio State University, Columbus, Ohio, United States of America
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, Ohio State University, Columbus, Ohio, United States of America
| | - Susan R. Mallery
- Division of Oral Pathology and Radiology, College of Dentistry, Ohio State University, Columbus, Ohio, United States of America
| | - Henry W. Fields
- Division of Orthodontics, College of Dentistry, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|