51
|
Periosteum and development of the tissue-engineered periosteum for guided bone regeneration. J Orthop Translat 2022; 33:41-54. [PMID: 35228996 PMCID: PMC8858911 DOI: 10.1016/j.jot.2022.01.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/02/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Background Periosteum plays a significant role in bone formation and regeneration by storing progenitor cells, and also acts as a source of local growth factors and a scaffold for recruiting cells and other growth factors. Recently, tissue-engineered periosteum has been studied extensively and shown to be important for osteogenesis and chondrogenesis. Using biomimetic methods for artificial periosteum synthesis, membranous tissues with similar function and structure to native periosteum are produced that significantly improve the efficacy of bone grafting and scaffold engineering, and can serve as direct replacements for native periosteum. Many problems involving bone defects can be solved by preparation of idealized periosteum from materials with different properties using various techniques. Methods This review summarizes the significance of periosteum for osteogenesis and chondrogenesis from the aspects of periosteum tissue structure, osteogenesis performance, clinical application, and development of periosteum tissue engineering. The advantages and disadvantages of different tissue engineering methods are also summarized. Results The fast-developing field of periosteum tissue engineering is aimed toward synthesis of bionic periosteum that can ensure or accelerate the repair of bone defects. Artificial periosteum materials can be similar to natural periosteum in both structure and function, and have good therapeutic potential. Induction of periosteum tissue regeneration and bone regeneration by biomimetic periosteum is the ideal process for bone repair. Conclusions Periosteum is essential for bone formation and regeneration, and it is indispensable in bone repair. Achieving personalized structure and composition in the construction of tissue engineering periosteum is in accordance with the design concept of both universality and emphasis on individual differences and ensures the combination of commonness and individuality, which are expected to meet the clinical needs of bone repair more effectively. The translational potential of this article To better understand the role of periosteum in bone repair, clarify the present research situation of periosteum and tissue engineering periosteum, and determine the development and optimization direction of tissue engineering periosteum in the future. It is hoped that periosteum tissue engineering will play a greater role in meeting the clinical needs of bone repair in the future, and makes it possible to achieve optimization of bone tissue therapy.
Collapse
|
52
|
Zhuang Y, Zhao Z, Cheng M, Li M, Si J, Lin K, Yu H. HIF-1α Regulates Osteogenesis of Periosteum-Derived Stem Cells Under Hypoxia Conditions via Modulating POSTN Expression. Front Cell Dev Biol 2022; 10:836285. [PMID: 35252198 PMCID: PMC8891937 DOI: 10.3389/fcell.2022.836285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/23/2022] Open
Abstract
Periosteum is indispensable in bone repair and is an important source of skeletal stem cells (SSCs) for endogenous bone regeneration. However, there are only a few studies about SSCs in periosteum. The craniomaxillofacial bone regeneration is done under the hypoxia microenvironment, in which HIF-1α plays an important role. The effect of HIF-1α on periosteum-derived stem cells (PDSCs) and the mechanisms of PDSCs activation under hypoxia conditions are unknown. In this study, the calvarial bone defect was established, with the periosteum removed or retained. Results show that the bone regeneration was severely impaired in the periosteum removed group. Moreover, pluripotent PDSCs isolated from the periosteum were positive for mesenchymal stem cell (MSC) markers. To determine the role of HIF-1α, the expression of HIF-1α was knocked down in vivo and in vitro, impairing the bone regeneration or osteogenesis of PDSCs. Furthermore, the knockdown of HIF-1α expression also reduced periostin (POSTN) expression, and recombinant POSTN addition partly rescued the osteogenic inhibition. Finally, to explore the mechanism under POSTN activation, the phosphorylation level of the PI3K/AKT pathway was assessed in transfected PDSCs. The phosphorylation level of PI3K and AKT was enhanced with HIF-1α overexpression and inhibited with HIF-1α knockdown, and the addition of PI3K activator or AKT activator could partly rescue POSTN expression. In conclusion, as a potential target to promote bone repair under the hypoxia microenvironment, HIF-1α can regulate the osteogenic differentiation of PDSCs via the PI3K/AKT/POSTN pathway, which lay a solid foundation for periosteum-based craniomaxillofacial bone regeneration.
Collapse
Affiliation(s)
- Yu Zhuang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhiyang Zhao
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Mengjia Cheng
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Meng Li
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiawen Si
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
- *Correspondence: Jiawen Si, ; Kaili Lin, ; Hongbo Yu,
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
- *Correspondence: Jiawen Si, ; Kaili Lin, ; Hongbo Yu,
| | - Hongbo Yu
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
- *Correspondence: Jiawen Si, ; Kaili Lin, ; Hongbo Yu,
| |
Collapse
|
53
|
On the horizon: Hedgehog signaling to heal broken bones. Bone Res 2022; 10:13. [PMID: 35165260 PMCID: PMC8844053 DOI: 10.1038/s41413-021-00184-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022] Open
Abstract
Uncovering the molecular pathways that drive skeletal repair has been an ongoing challenge. Initial efforts have relied on in vitro assays to identify the key signaling pathways that drive cartilage and bone differentiation. While these assays can provide some clues, assessing specific pathways in animal models is critical. Furthermore, definitive proof that a pathway is required for skeletal repair is best provided using genetic tests. Stimulating the Hh (Hedgehog) pathway can promote cartilage and bone differentiation in cell culture assays. In addition, the application of HH protein or various pathway agonists in vivo has a positive influence on bone healing. Until recently, however, genetic proof that the Hh pathway is involved in bone repair has been lacking. Here, we consider both in vitro and in vivo studies that examine the role of Hh in repair and discuss some of the challenges inherent in their interpretation. We also identify needed areas of study considering a new appreciation for the role of cartilage during repair, the variety of cell types that may have differing roles in repair, and the recent availability of powerful lineage tracing techniques. We are optimistic that emerging genetic tools will make it possible to precisely define when and in which cells promoting Hh signaling can best promote skeletal repair, and thus, the clinical potential for targeting the Hh pathway can be realized.
Collapse
|
54
|
Targeted activation of androgen receptor signaling in the periosteum improves bone fracture repair. Cell Death Dis 2022; 13:123. [PMID: 35136023 PMCID: PMC8826926 DOI: 10.1038/s41419-022-04595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/30/2021] [Accepted: 01/27/2022] [Indexed: 12/03/2022]
Abstract
Low testosterone level is an independent predictor of osteoporotic fracture in elderly men as well as increased fracture risk in men undergoing androgen deprivation. Androgens and androgen receptor (AR) actions are essential for bone development and homeostasis but their linkage to fracture repair remains unclear. Here we found that AR is highly expressed in the periosteum cells and is co-localized with a mesenchymal progenitor cell marker, paired-related homeobox protein 1 (Prrx1), during bone fracture repair. Mice lacking the AR gene in the periosteum expressing Prrx1-cre (AR-/Y;Prrx1::Cre) but not in the chondrocytes (AR-/Y;Col-2::Cre) exhibits reduced callus size and new bone volume. Gene expression data analysis revealed that the expression of several collagens, integrins and cell adhesion molecules were downregulated in periosteum-derived progenitor cells (PDCs) from AR-/Y;Prrx1::Cre mice. Mechanistically, androgens-AR signaling activates the AR/ARA55/FAK complex and induces the collagen-integrin α2β1 gene expression that is required for promoting the AR-mediated PDCs migration. Using mouse cortical-defect and femoral graft transplantation models, we proved that elimination of AR in periosteum of host mice impairs fracture healing, regardless of AR existence of transplanted donor graft. While testosterone implanted scaffolds failed to complete callus bridging across the fracture gap in AR-/Y;Prrx1::Cre mice, cell-based transplantation using DPCs re-expressing AR could lead to rescue bone repair. In conclusion, targeting androgen/AR axis in the periosteum may provide a novel therapy approach to improve fracture healing.
Collapse
|
55
|
Case series of congenital pseudarthrosis of the tibia unfulfilling neurofibromatosis type 1 diagnosis: 21% with somatic NF1 haploinsufficiency in the periosteum. Hum Genet 2022; 141:1371-1383. [DOI: 10.1007/s00439-021-02429-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/29/2021] [Indexed: 12/17/2022]
|
56
|
Qi T, Li L, Weidong T. The Role of Sphingolipid Metabolism in Bone Remodeling. Front Cell Dev Biol 2021; 9:752540. [PMID: 34912800 PMCID: PMC8666436 DOI: 10.3389/fcell.2021.752540] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/11/2021] [Indexed: 02/05/2023] Open
Abstract
Emerging studies of bioactive lipids have made many exciting discoveries in recent years. Sphingolipids and their metabolites perform a wide variety of cellular functions beyond energy metabolism. Emerging evidence based on genetically manipulated mouse models and molecular biology allows us to obtain new insights into the role sphingolipid played on skeletal remodeling. This review summarizes studies or understandings of the crosstalk between sphingomyelin, ceramide, and sphingosine-1-phosphate (S1P) of sphingolipids family and the cells, especially osteoblasts and osteoclasts of the bone through which bone is remodeled during life constantly. This review also shows agonists and antagonists of S1P as possible therapeutic options and opportunities on bone diseases.
Collapse
Affiliation(s)
- Tang Qi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Liao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tian Weidong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
57
|
Akiyama M. Role of FBXW2 in explant cultures of bovine periosteum-derived cells. BMC Res Notes 2021; 14:410. [PMID: 34736516 PMCID: PMC8569954 DOI: 10.1186/s13104-021-05825-z] [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: 07/21/2021] [Accepted: 10/26/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Bone regeneration is a potential technique for treating osteoporosis. A previous study reported that F-box and WD-40 domain-containing protein 2 (FBXW2) localized with osteocalcin in bovine periosteum after 5 weeks of explant culture. However, the osteoblastic functions of FBXW2 remain unclear. In this study, double-fluorescent immunostaining was used to investigate the potential role of FBXW2 and its relationship with osteocalcin. RESULTS At day 0, FBXW2 was expressed in the cambium layer between the bone and periosteum, while osteocalcin was expressed in bone. After explant culture, changes in the periosteum were observed from weeks 1 to 7. At week 1, partial FBXW2 expression was seen with a small amount of osteocalcin. At week 2, a layer of FBXW2 was observed. From weeks 3 to 7, tube-like structures of FBXW and osteocalcin were observed, and periosteum-derived cells were released from the periosteum in areas where no FBXW2 was observed. Bovine periosteum-derived cells can form a three-dimensional cell pellet, because multilayered cell sheets are formed inside of the periosteum in vitro. It is shown that in results FBXW2 is produced in periosteal explants near sites where initial osteogenic activity is observed, suggesting that it may be involved in periosteal osteogenesis.
Collapse
Affiliation(s)
- Mari Akiyama
- Department of Biomaterials, Osaka Dental University, 8-1 Kuzuhahanozono-cho, Hirakata-shi, Osaka, 573-1121, Japan.
| |
Collapse
|
58
|
Influence of Human Jaw Periosteal Cells Seeded β-Tricalcium Phosphate Scaffolds on Blood Coagulation. Int J Mol Sci 2021; 22:ijms22189942. [PMID: 34576103 PMCID: PMC8467579 DOI: 10.3390/ijms22189942] [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: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering offers auspicious opportunities in oral and maxillofacial surgery to heal bone defects. For this purpose, the combination of cells with stability-providing scaffolds is required. Jaw periosteal cells (JPCs) are well suited for regenerative therapies, as they are easily accessible and show strong osteogenic potential. In this study, we analyzed the influence of uncoated and polylactic-co-glycolic acid (PLGA)-coated β-tricalcium phosphate (β-TCP) scaffolds on JPC colonization and subsequent osteogenic differentiation. Furthermore, interaction with the human blood was investigated. This study demonstrated that PLGA-coated and uncoated β-TCP scaffolds can be colonized with JPCs and further differentiated into osteogenic cells. On day 15, after cell seeding, JPCs with and without osteogenic differentiation were incubated with fresh human whole blood under dynamic conditions. The activation of coagulation, complement system, inflammation, and blood cells were analyzed using ELISA and scanning electron microscopy (SEM). JPC-seeded scaffolds showed a dense cell layer and osteogenic differentiation capacity on both PLGA-coated and uncoated β-TCP scaffolds. SEM analyses showed no relevant blood cell attachment and ELISA results revealed no significant increase in most of the analyzed cell activation markers (β-thromboglobulin, Sc5B-9, polymorphonuclear (PMN)-elastase). However, a notable increase in thrombin-antithrombin III (TAT) complex levels, as well as fibrin fiber accumulation on JPC-seeded β-TCP scaffolds, was detected compared to the scaffolds without JPCs. Thus, this study demonstrated that besides the scaffold material the cells colonizing the scaffolds can also influence hemostasis, which can influence the regeneration of bone tissue.
Collapse
|
59
|
Liu YL, Yen CC, Liu TST, Chang CH, Shih TTF, Wang JH, Yang MC, Lin FH, Liu HC. Safety and Efficacy of Kartigen ® in Treating Cartilage Defects: A Randomized, Controlled, Phase I Trial. Polymers (Basel) 2021; 13:polym13183029. [PMID: 34577930 PMCID: PMC8466236 DOI: 10.3390/polym13183029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 01/18/2023] Open
Abstract
Here, we aimed to investigate the safety and preliminary efficacy of Kartigen®, a matrix with autologous bone marrow mesenchymal stem cell-derived chondrocyte precursors embedded in atelocollagen. As a surgical graft, Kartigen® was implanted onto the cartilage defects at the weight-bearing site of the medial femoral condyle of the knee. Fifteen patients were enrolled and stratified into two groups, undergoing either Kartigen® implantation (n = 10) or microfracture (control group, n = 5). The primary endpoint was to evaluate the safety of Kartigen® by monitoring the occurrence of adverse events through physician queries, physical examinations, laboratory tests, and radiological analyses for 2 years. There were no infections, inflammations, adhesions, loose body, or tumor formations in the Kartigen®-implanted knees. The preliminary efficacy was assessed using the International Knee Documentation Committee (IKDC) score, visual analog scale, and second-look arthroscopy. The postoperative IKDC scores of the Kartigen® group significantly improved in the 16th week (IKDC = 62.1 ± 12.8, p = 0.025), kept increasing in the first year (IKDC = 78.2 ± 15.4, p < 0.005), and remained satisfactory in the second year (IKDC = 73.6 ± 13.8, p < 0.005), compared to the preoperative condition (IKDC = 47.1 ± 17.0), while the postoperative IKDC scores of the control group also achieved significant improvement in the 28th week (IKDC = 68.5 ± 6.1, p = 0.032) versus preoperative state (IKDC = 54.0 ± 9.1). However, the IKDC scores decreased in the first year (IKDC = 63.5 ± 11.6) as well as in the second year (IKDC = 52.6 ± 16.4). Thirteen patients underwent second-look arthroscopy and biopsy one year after the operation. The Kartigen® group exhibited integration between Kartigen® and host tissue with a smooth appearance at the recipient site, whereas the microfracture group showed fibrillated surfaces. The histological and immunohistochemical analyses of biopsy specimens demonstrated the columnar structure of articular cartilage and existence of collagen type II and glycosaminoglycan mimic hyaline cartilage. This study indicates that Kartigen® is safe and effective in treating cartilage defects.
Collapse
Affiliation(s)
- Yen-Liang Liu
- Master Program for Biomedical Engineering, College of Biomedical Engineering, China Medical University, Taichung 406040, Taiwan;
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Chun-Che Yen
- Kartigen Biomedical Inc., Taipei 100047, Taiwan;
| | | | - Chih-Hung Chang
- Department of Orthopaedic Surgery, Far Eastern Memorial Hospital, New Taipei 220216, Taiwan;
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| | - Tiffany Ting-Fang Shih
- Department of Medical Imaging and Radiology, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Jyh-Horng Wang
- Department of Orthopaedic Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Ming-Chia Yang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 310401, Taiwan;
| | - Feng-Huei Lin
- Department of Biomedical Engineering, College of Engineering, National Taiwan University, Taipei 106319, Taiwan;
| | - Hwa-Chang Liu
- Department of Orthopaedic Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan;
- Department of Orthopaedic Surgery, Taiwan Adventist Hospital, Taipei 105404, Taiwan
- Correspondence:
| |
Collapse
|
60
|
Free Periosteal Flaps with Scaffold: An Overlooked Armamentarium for Maxillary and Mandibular Reconstruction. Cancers (Basel) 2021; 13:cancers13174373. [PMID: 34503183 PMCID: PMC8431391 DOI: 10.3390/cancers13174373] [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: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary Head and neck bone reconstruction with revascularized free periosteal flaps and scaffold is an overlooked option in the literature. Aim of the present paper was to systematically analyse the results of maxillary and mandibular reconstruction with this technique. We found a total of 7 studies with 55 patients fitting with our inclusion criteria. The overall rate of complications was 43.7%. The success rate intended as scaffold integration resulted to be 74.5%. Our paper therefore highlighted that maxillary and mandibular reconstruction with revascularized free periosteal flaps and scaffold is a possible alternative in patient unable to bone free flap complex reconstruction, with a success rate higher to that of other secondary options. Abstract Introduction: Head and neck bone reconstruction is a challenging surgical scenario. Although several strategies have been described in the literature, bone free flaps (BFFs) have become the preferred technique for large defects. Revascularized free periosteal flaps (FPFs) with support scaffold represents a possible alternative in compromised patient, BFF failure, or relapsing cancers as salvage treatment. However, only few clinical applications in head and neck are reported in literature. Purpose of the study was to systematically analyse the results of functional and oncologic maxillary and mandibular reconstruction with FPF with scaffold. Materials and Methods: A comprehensive review of the dedicated literature was performed according to the PRISMA guidelines searching on Scopus, PubMed/MEDLINE, Cochrane Library, Embase, Researchgate and Google Scholar databases using relevant keywords, phrases and medical subject headings (MeSH) terms. An excursus on the most valuable FPF’ harvesting sites was also carried out. Results: A total of 7 studies with 55 patients were included. Overall, the majority of the patients (n = 54, 98.1%) underwent an FPF reconstruction of the mandibular site. The most used technique was the radial forearm FPF with autologous frozen bone as scaffold (n = 40, 72.7%). The overall rate of complications was 43.7%. The success rate intended as scaffold integration resulted to be 74.5%. Conclusions: Maxillary and mandibular reconstruction with FPF and scaffold is a possible alternative in patient unfit for complex BFF reconstruction and it should be considered as a valid alternative in the sequential salvage surgery for locally advanced cancer. Moreover, it opens future scenarios in head and neck reconstructive surgery, as a promising tool that can be modelled to tailor complex 3D defects, with less morbidities to the donor site.
Collapse
|
61
|
Moncal KK, Gudapati H, Godzik KP, Heo DN, Kang Y, Rizk E, Ravnic DJ, Wee H, Pepley DF, Ozbolat V, Lewis GS, Moore JZ, Driskell RR, Samson TD, Ozbolat IT. Intra-Operative Bioprinting of Hard, Soft, and Hard/Soft Composite Tissues for Craniomaxillofacial Reconstruction. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2010858. [PMID: 34421475 PMCID: PMC8376234 DOI: 10.1002/adfm.202010858] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Indexed: 05/20/2023]
Abstract
Reconstruction of complex craniomaxillofacial (CMF) defects is challenging due to the highly organized layering of multiple tissue types. Such compartmentalization necessitates the precise and effective use of cells and other biologics to recapitulate the native tissue anatomy. In this study, intra-operative bioprinting (IOB) of different CMF tissues, including bone, skin, and composite (hard/soft) tissues, is demonstrated directly on rats in a surgical setting. A novel extrudable osteogenic hard tissue ink is introduced, which induced substantial bone regeneration, with ≈80% bone coverage area of calvarial defects in 6 weeks. Using droplet-based bioprinting, the soft tissue ink accelerated the reconstruction of full-thickness skin defects and facilitated up to 60% wound closure in 6 days. Most importantly, the use of a hybrid IOB approach is unveiled to reconstitute hard/soft composite tissues in a stratified arrangement with controlled spatial bioink deposition conforming the shape of a new composite defect model, which resulted in ≈80% skin wound closure in 10 days and 50% bone coverage area at Week 6. The presented approach will be absolutely unique in the clinical realm of CMF defects and will have a significant impact on translating bioprinting technologies into the clinic in the future.
Collapse
Affiliation(s)
- Kazim K Moncal
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hemanth Gudapati
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kevin P Godzik
- Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Dong N Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Youngnam Kang
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Elias Rizk
- Department of Neurosurgery, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Dino J Ravnic
- Department of Surgery, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Hwabok Wee
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - David F Pepley
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Veli Ozbolat
- Mechanical Engineering Department, Ceyhan Engineering Faculty, Cukurova University, Adana 01950, Turkey
| | - Gregory S Lewis
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jason Z Moore
- Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Ryan R Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Thomas D Samson
- Department of Neurosurgery, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
62
|
Atelocollagen-Embedded Chondrocyte Precursors as a Treatment for Grade-4 Cartilage Defects of the Femoral Condyle: A Case Series with up to 9-Year Follow-Up. Biomolecules 2021; 11:biom11070942. [PMID: 34202002 PMCID: PMC8301975 DOI: 10.3390/biom11070942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 12/22/2022] Open
Abstract
We demonstrated the safety and efficacy of autologous chondrocyte precursor (CP) cell therapy in repairing Grade 4 cartilage defects of medial femoral condyles. The autologous bone marrow mesenchymal stem cells of each participant were isolated, amplified, and then differentiated into CPs in atelocollagen. Neotissues made of CPs were implanted into cartilage defects with an average cell density of 4.9 ± 2.1 × 106 cells/cm2 through arthrotomy. The knee function was evaluated with the International Knee Documentation Committee (IKDC) subjective knee form. Patients' knee functions significantly improved by the 28th week (IKDC score = 68.3 ± 12.1), relative to the initial functionality before the CP therapy (IKDC score = 46.1 ± 16.4, p-value = 0.0014). Nine of these twelve patients maintained good knee functions for 9 years post-implantation (IKDC score = 69.8 ± 12.3) at levels higher than the pre-implantation values (p-value = 0.0018). Patients were evaluated with MRI and arthroscopy, and the defective sites exhibited a smooth surface without a gap between the implant and host tissue. This study demonstrates that autologous CPs successfully engraft into the host tissue and result in the re-formation of hyaline-like cartilage, thereby improving the impaired knee functions. Most importantly, no adverse event was reported during this long-term follow-up period.
Collapse
|
63
|
Shah M, Maroof A, Gikas P, Mittal G, Keen R, Baeten D, Shaw S, Roberts SJ. Dual neutralisation of IL-17F and IL-17A with bimekizumab blocks inflammation-driven osteogenic differentiation of human periosteal cells. RMD Open 2021; 6:rmdopen-2020-001306. [PMID: 32723833 PMCID: PMC7722278 DOI: 10.1136/rmdopen-2020-001306] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/25/2020] [Accepted: 06/25/2020] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES Interleukin (IL)-17 signalling has been shown to be a key regulator of disease in ankylosing spondylitis (AS) with several IL-17 blockers currently clinically approved. Despite this, the role of IL-17 in bone pathology is poorly understood. This study aimed to investigate IL-17 signalling in the context of pathological bone formation. METHODS A biomimetic human periosteum-derived cell (hPDC) model of osteogenic differentiation was used in combination with recombinant IL-17 cytokines, T-cell supernatants or serum from patients with AS. IL-17A, IL-17F and bimekizumab monoclonal antibodies were used to block IL-17 cytokine action. RESULTS Recombinant IL-17A and IL-17F are pro-osteogenic with respect to hPDC differentiation. T helper 17 or γδ-T cell supernatants also potently stimulated in vitro bone formation, which was blocked deeper by dual inhibition of IL-17A and IL-17F than by neutralisation of IL-17A or IL-17F individually. Osteogenic blockade may be due to an increase in expression of the Wnt antagonist DKK1. Interestingly, osteocommitment was also induced by serum obtained from patients with AS, which was also abrogated by dual neutralisation of IL-17A and IL-17F. CONCLUSIONS These data show for the first time that IL-17A and IL-17F enhance in vitro osteogenic differentiation and bone formation from hPDCs, inhibition of which may offer an attractive therapeutic strategy to prevent pathological bone formation.
Collapse
Affiliation(s)
- Mittal Shah
- UCB Pharma, Slough, UK.,Division of Surgery and Interventional Science, University College London, London, UK
| | | | - Panos Gikas
- Royal National Orthopaedic Hospital Stanmore, Stanmore, UK
| | - Gayatri Mittal
- Royal National Orthopaedic Hospital Stanmore, Stanmore, UK
| | - Richard Keen
- Royal National Orthopaedic Hospital Stanmore, Stanmore, UK
| | | | | | - Scott J Roberts
- UCB Pharma, Slough, UK .,Division of Surgery and Interventional Science, University College London, London, UK.,Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
64
|
Lou Y, Wang H, Ye G, Li Y, Liu C, Yu M, Ying B. Periosteal Tissue Engineering: Current Developments and Perspectives. Adv Healthc Mater 2021; 10:e2100215. [PMID: 33938636 DOI: 10.1002/adhm.202100215] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/18/2021] [Indexed: 12/22/2022]
Abstract
Periosteum, a highly vascularized bilayer connective tissue membrane plays an indispensable role in the repair and regeneration of bone defects. It is involved in blood supply and delivery of progenitor cells and bioactive molecules in the defect area. However, sources of natural periosteum are limited, therefore, there is a need to develop tissue-engineered periosteum (TEP) mimicking the composition, structure, and function of natural periosteum. This review explores TEP construction strategies from the following perspectives: i) different materials for constructing TEP scaffolds; ii) mechanical properties and surface topography in TEP; iii) cell-based strategies for TEP construction; and iv) TEP combined with growth factors. In addition, current challenges and future perspectives for development of TEP are discussed.
Collapse
Affiliation(s)
- Yiting Lou
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Guanchen Ye
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Yongzheng Li
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Chao Liu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Binbin Ying
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| |
Collapse
|
65
|
Wang J, Wang X, Zhen P, Fan B. [Research progress of in vivo bioreactor for bone tissue engineering]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:627-635. [PMID: 33998218 DOI: 10.7507/1002-1892.202012083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To review the research progress of in vivo bioreactor (IVB) for bone tissue engineering in order to provide reference for its future research direction. Methods The literature related to IVB used in bone tissue engineering in recent years was reviewed, and the principles of IVB construction, tissue types, sites, and methods of IVB construction, as well as the advantages of IVB used in bone tissue engineering were summarized. Results IVB takes advantage of the body's ability to regenerate itself, using the body as a bioreactor to regenerate new tissues or organs at injured sites or at ectopic sites that can support the regeneration of new tissues. IVB can be constructed by tissue flap (subcutaneous pocket, muscle flap/pocket, fascia flap, periosteum flap, omentum flap/abdominal cavity) and axial vascular pedicle (axial vascular bundle, arteriovenous loop) alone or jointly. IVB is used to prefabricate vascularized tissue engineered bone that matched the shape and size of the defect. The prefabricated vascularized tissue engineered bone can be used as bone graft, pedicled bone flap, or free bone flap to repair bone defect. IVB solves the problem of insufficient vascularization in traditional bone tissue engineering to a certain extent. Conclusion IVB is a promising method for vascularized tissue engineered bone prefabrication and subsequent bone defect reconstruction, with unique advantages in the repair of large complex bone defects. However, the complexity of IVB construction and surgical complications hinder the clinical application of IVB. Researchers should aim to develop a simple, safe, and efficient IVB.
Collapse
Affiliation(s)
- Jian Wang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou Gansu, 730000, P.R.China.,Orthopaedic Center, the 940th Hospital of PLA Joint Logistics Support Force, Lanzhou Gansu, 730000, P.R.China
| | - Xiao Wang
- School of Design and Art, Lanzhou University of Technology, Lanzhou Gansu, 730000, P.R.China
| | - Ping Zhen
- Orthopaedic Center, the 940th Hospital of PLA Joint Logistics Support Force, Lanzhou Gansu, 730000, P.R.China
| | - Bo Fan
- Orthopaedic Center, the 940th Hospital of PLA Joint Logistics Support Force, Lanzhou Gansu, 730000, P.R.China
| |
Collapse
|
66
|
Ishizuka S, Dong QN, Ngo HX, Bai Y, Sha J, Toda E, Okui T, Kanno T. Bioactive Regeneration Potential of the Newly Developed Uncalcined/Unsintered Hydroxyapatite and Poly-l-Lactide-Co-Glycolide Biomaterial in Maxillofacial Reconstructive Surgery: An In Vivo Preliminary Study. MATERIALS 2021; 14:ma14092461. [PMID: 34068558 PMCID: PMC8126161 DOI: 10.3390/ma14092461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022]
Abstract
Uncalcined/unsintered hydroxyapatite (HA) and poly-l-lactide-co-glycolide (u-HA/PLLA/PGA) are novel bioresorbable bioactive materials with bone regeneration characteristics and have been used to treat mandibular defects in a rat model. However, the bone regenerative interaction with the periosteum, the inflammatory response, and the degradation of this material have not been examined. In this study, we used a rat mandible model to compare the above features in u-HA/PLLA/PGA and uncalcined/unsintered HA and poly-l-lactic acid (u-HA/PLLA). We divided 11 male Sprague–Dawley rats into 3- and 16-week groups. In each group, we assessed the characteristics of a u-HA/PLLA/PGA sheet covering the right mandibular angle and a u-HA/PLLA sheet covering the left mandibular angle in three rats each, and one rat was used as a sham control. The remaining three rats in the 16-week group were used for a degradation assessment and received both sheets of material as in the material assessment subgroup. At 3 and 16 weeks after surgery, the rats were sacrificed, and mandible specimens were subjected to micro-computed tomography, histological analysis, and immunohistochemical staining. The results indicated that the interaction between the periosteum and u-HA/PLLA/PGA material produced significantly more new bone regeneration with a lower inflammatory response and a faster resorption rate compared to u-HA/PLLA alone. These findings may indicate that this new biomaterial has ideal potential in treating maxillofacial defects of the midface and orbital regions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Takahiro Kanno
- Correspondence: ; Tel.: +81-(0)853-20-2301; Fax: +81-(0)853-20-2299
| |
Collapse
|
67
|
Shiferaw YF, Engidaw MT, Kedir M, Tsegaye AT. Wealth index is significantly associated with the early phase of fracture healing among fractured patients at the University of Gondar Specialized Hospital, Northwest Ethiopia. J Public Health (Oxf) 2021. [DOI: 10.1007/s10389-021-01559-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
68
|
Xiao H, Chen J, Duan L, Li S. Role of emerging vitamin K‑dependent proteins: Growth arrest‑specific protein 6, Gla‑rich protein and periostin (Review). Int J Mol Med 2021; 47:2. [PMID: 33448308 PMCID: PMC7834955 DOI: 10.3892/ijmm.2020.4835] [Citation(s) in RCA: 15] [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: 04/13/2020] [Accepted: 10/21/2020] [Indexed: 01/27/2023] Open
Abstract
Vitamin K‑dependent proteins (VKDPs) are a group of proteins that need vitamin K to conduct carboxylation. Thus far, scholars have identified a total of 17 VKDPs in the human body. In this review, we summarize three important emerging VKDPs: Growth arrest‑specific protein 6 (Gas 6), Gla‑rich protein (GRP) and periostin in terms of their functions in physiological and pathological conditions. As examples, carboxylated Gas 6 and GRP effectively protect blood vessels from calcification, Gas 6 protects from acute kidney injury and is involved in chronic kidney disease, GRP contributes to bone homeostasis and delays the progression of osteoarthritis, and periostin is involved in all phases of fracture healing and assists myocardial regeneration in the early stages of myocardial infarction. However, periostin participates in the progression of cardiac fibrosis, idiopathic pulmonary fibrosis and airway remodeling of asthma. In addition, we discuss the relationship between vitamin K, VKDPs and cancer, and particularly the carboxylation state of VKDPs in cancer.
Collapse
Affiliation(s)
- Huiyu Xiao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| | - Jiepeng Chen
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Lili Duan
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| |
Collapse
|
69
|
Matthews BG, Novak S, Sbrana FV, Funnell JL, Cao Y, Buckels EJ, Grcevic D, Kalajzic I. Heterogeneity of murine periosteum progenitors involved in fracture healing. eLife 2021; 10:e58534. [PMID: 33560227 PMCID: PMC7906599 DOI: 10.7554/elife.58534] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
The periosteum is the major source of cells involved in fracture healing. We sought to characterize progenitor cells and their contribution to bone fracture healing. The periosteum is highly enriched with progenitor cells, including Sca1+ cells, fibroblast colony-forming units, and label-retaining cells compared to the endosteum and bone marrow. Using lineage tracing, we demonstrate that alpha smooth muscle actin (αSMA) identifies long-term, slow-cycling, self-renewing osteochondroprogenitors in the adult periosteum that are functionally important for bone formation during fracture healing. In addition, Col2.3CreER-labeled osteoblast cells contribute around 10% of osteoblasts but no chondrocytes in fracture calluses. Most periosteal osteochondroprogenitors following fracture can be targeted by αSMACreER. Previously identified skeletal stem cell populations were common in periosteum but contained high proportions of mature osteoblasts. We have demonstrated that the periosteum is highly enriched with skeletal progenitor cells, and there is heterogeneity in the populations of cells that contribute to mature lineages during periosteal fracture healing.
Collapse
Affiliation(s)
- Brya G Matthews
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Sanja Novak
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Francesca V Sbrana
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Jessica L Funnell
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Ye Cao
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
| | - Emma J Buckels
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
| | - Danka Grcevic
- Department of Physiology and Immunology, University of ZagrebZagrebCroatia
- Croatian Intitute for Brain Research, University of ZagrebZagrebCroatia
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| |
Collapse
|
70
|
Doherty L, Wan M, Kalajzic I, Sanjay A. Diabetes impairs periosteal progenitor regenerative potential. Bone 2021; 143:115764. [PMID: 33221502 PMCID: PMC7770068 DOI: 10.1016/j.bone.2020.115764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
Diabetics are at increased risk for fracture, and experience severely impaired skeletal healing characterized by delayed union or nonunion of the bone. The periosteum harbors osteochondral progenitors that can differentiate into chondrocytes and osteoblasts, and this connective tissue layer is required for efficient fracture healing. While bone marrow-derived stromal cells have been studied extensively in the context of diabetic skeletal repair and osteogenesis, the effect of diabetes on the periosteum and its ability to contribute to bone regeneration has not yet been explicitly evaluated. Within this study, we utilized an established murine model of type I diabetes to evaluate periosteal cell differentiation capacity, proliferation, and availability under the effect of a diabetic environment. Periosteal cells from diabetic mice were deficient in osteogenic differentiation ability in vitro, and diabetic mice had reduced periosteal populations of mesenchymal progenitors with a corresponding reduction in proliferation capacity following injury. Additionally, fracture callus mineralization and mature osteoblast activity during periosteum-mediated healing was impaired in diabetic mice compared to controls. We propose that the effect of diabetes on periosteal progenitors and their ability to aid in skeletal repair directly impairs fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn School of Dental Medicine, Farmington, CT, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| |
Collapse
|
71
|
Miyazaki-Asato Y, Koi K, Fujimoto H, Kakura K, Kido H, Yanagi T, Yamashita J. Intramedullary injury combined with osteoporosis therapeutics regulates targeted local osteogenesis. Sci Rep 2021; 11:205. [PMID: 33436871 PMCID: PMC7804436 DOI: 10.1038/s41598-020-80316-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022] Open
Abstract
Bone marrow ablation prompts transient bone formation in nearly the entire medullary cavity before marrow regeneration occurs. Here, we establish a procedure to direct bone formation in a desired particular site within the medullary cavity for support of biomedical devices. Local intramedullary injury was performed in the tibiae of rats and parathyroid hormone (PTH), alendronate, or saline was administered. Newly generated bone in the medulla was assessed by micro-CT and histology. To evaluate the function of newly generated bone, animals received intramedullary injury in tibiae followed by daily PTH. At day-14, implants were placed in the endocortical bone and the bone response to the implants was assessed. The fate of newly generated bone was compared with and without implants. We found that neither intramedullary injury nor medication alone resulted in bone formation. However, when combined, substantial bone was generated locally inside the diaphyseal medulla. Newly formed bone disappeared without implant placement but was retained with implants. Bone was especially retained around and between the implants. This study found that local bone marrow disruption followed by PTH or alendronate generated substantial cancellous bone locally in the diaphyseal medulla. This approach offers promise as a tissue engineering tool in medicine and dentistry.
Collapse
Affiliation(s)
- Yoko Miyazaki-Asato
- Department of Oral Rehabilitation, Oral Implantology, Fukuoka Dental College, Fukuoka, Japan
| | - Kiyono Koi
- Department of Restorative Dentistry, Oregon Health & Science University School of Dentistry, Portland, OR, USA
| | - Hiroki Fujimoto
- Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| | - Kae Kakura
- Department of Oral Rehabilitation, Oral Implantology, Fukuoka Dental College, Fukuoka, Japan
| | - Hirofumi Kido
- Department of Oral Rehabilitation, Oral Implantology, Fukuoka Dental College, Fukuoka, Japan
| | - Tsukasa Yanagi
- Department of Oral Rehabilitation, Oral Implantology, Fukuoka Dental College, Fukuoka, Japan
| | - Junro Yamashita
- Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan.
- Center for Regenerative Medicine, Fukuoka Dental College, Sawara-ku Tamura 2-15-1, Fukuoka, 814-0193, Japan.
| |
Collapse
|
72
|
van Gastel N, Carmeliet G. Metabolic regulation of skeletal cell fate and function in physiology and disease. Nat Metab 2021; 3:11-20. [PMID: 33398192 DOI: 10.1038/s42255-020-00321-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 01/28/2023]
Abstract
The skeleton is diverse in its functions, which include mechanical support, movement, blood cell production, mineral storage and endocrine regulation. This multifaceted role is achieved through an interplay of osteoblasts, chondrocytes, bone marrow adipocytes and stromal cells, all generated from skeletal stem cells. Emerging evidence shows the importance of cellular metabolism in the molecular control of the skeletal system. The different skeletal cell types not only have distinct metabolic demands relating to their particular functions but also are affected by microenvironmental constraints. Specific metabolites control skeletal stem cell maintenance, direct lineage allocation and mediate cellular communication. Here, we discuss recent findings on the roles of cellular metabolism in determining skeletal stem cell fate, coordinating osteoblast and chondrocyte function, and organizing stromal support of haematopoiesis. We also consider metabolic dysregulation in skeletal ageing and degenerative diseases, and provide an outlook on how the field may evolve in the coming years.
Collapse
Affiliation(s)
- Nick van Gastel
- de Duve Institute, Brussels, Belgium.
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium.
| |
Collapse
|
73
|
Naujokat H, Loger K, Schulz J, Açil Y, Wiltfang J. Bone tissue engineering in the greater omentum with computer-aided design/computer-aided manufacturing scaffolds is enhanced by a periosteum transplant. Regen Med 2020; 15:2297-2309. [PMID: 33355523 DOI: 10.2217/rme-2020-0115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: This study aimed to evaluate two different vascularized bone flap scaffolds and the impact of two barrier membranes for the reconstruction of critical-size bone defects. Materials & methods: 3D-printed scaffolds of biodegradable calcium phosphate and bioinert titanium were loaded with rhBMP-2 bone marrow aspirate, wrapped by a collagen membrane or a periosteum transplant and implanted into the greater omentum of miniature pigs. Results: Histological evaluation demonstrated significant bone formation within the first 8 weeks in both scaffolds. The periosteum transplant led to enhanced bone formation and a homogenous distribution in the scaffolds. The omentum tissue grew out a robust vascular supply. Conclusion: Endocultivation using 3D-printed scaffolds in the greater omentum is a very promising approach in defect-specific bone tissue regeneration.
Collapse
Affiliation(s)
- Hendrik Naujokat
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Klaas Loger
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Juliane Schulz
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Yahya Açil
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Jörg Wiltfang
- Department of Oral & Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| |
Collapse
|
74
|
Jin Y, Sun X, Pei F, Zhao Z, Mao J. Wnt16 signaling promotes osteoblast differentiation of periosteal derived cells in vitro and in vivo. PeerJ 2020; 8:e10374. [PMID: 33282557 PMCID: PMC7694570 DOI: 10.7717/peerj.10374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Background Periosteum plays critical roles in de novo bone formation and fracture repair. Wnt16 has been regarded as a key regulator in periosteum bone formation. However, the role of Wnt16 in periosteum derived cells (PDCs) osteogenic differentiation remains unclear. The study goal is to uncover whether and how Wnt16 acts on the osteogenesis of PDCs. Methods We detected the variation of Wnt16 mRNA expression in PDCs, which were isolated from mouse femur and identified by flow cytometry, cultured in osteogenic medium for 14 days, then knocked down and over-expressed Wnt16 in PDCs to analysis its effects in osteogenesis. Further, we seeded PDCs (Wnt16 over-expressed/vector) in β-tricalcium phosphate cubes, and transplanted this complex into a critical size calvarial defect. Lastly, we used immunofluorescence, Topflash and NFAT luciferase reporter assay to study the possible downstream signaling pathway of Wnt16. Results Wnt16 mRNA expression showed an increasing trend in PDCs under osteogenic induction for 14 days. Wnt16 shRNA reduced mRNA expression of Runx2, collage type I (Col-1) and osteocalcin (OCN) after 7 days of osteogenic induction, as well as alizarin red staining intensity after 21days. Wnt16 also increased the mRNA expression of Runx2 and OCN and the protein production of Runx2 and Col-1 after 2 days of osteogenic stimulation. In the orthotopic transplantation assay, more bone volume, trabecula number and less trabecula space were found in Wnt16 over-expressed group. Besides, in the newly formed tissue Brdu positive area was smaller and Col-1 was larger in Wnt16 over-expressed group compared to the control group. Finally, Wnt16 upregulated CTNNB1/β-catenin expression and its nuclear translocation in PDCs, also increased Topflash reporter luciferase activity. By contrast, Wnt16 failed to increase NFAT reporter luciferase activity. Conclusion Together, Wnt16 plays a positive role in regulating PDCs osteogenesis, and Wnt16 may have a potential use in improving bone regeneration.
Collapse
Affiliation(s)
- Ying Jin
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyan Sun
- Stomatological Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Pei
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jeremy Mao
- Columbia University, Center for Craniofacial Regeneration, New York, NY, United States of America
| |
Collapse
|
75
|
Groeneveldt LC, Herpelinck T, Maréchal M, Politis C, van IJcken WFJ, Huylebroeck D, Geris L, Mulugeta E, Luyten FP. The Bone-Forming Properties of Periosteum-Derived Cells Differ Between Harvest Sites. Front Cell Dev Biol 2020; 8:554984. [PMID: 33324630 PMCID: PMC7723972 DOI: 10.3389/fcell.2020.554984] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
The development of alternatives for autologous bone grafts is a major focus of bone tissue engineering. To produce living bone-forming implants, skeletal stem and progenitor cells (SSPCs) are envisioned as key ingredients. SSPCs can be obtained from different tissues including bone marrow, adipose tissue, dental pulp, and periosteum. Human periosteum-derived cells (hPDCs) exhibit progenitor cell characteristics and have well-documented in vivo bone formation potency. Here, we have characterized and compared hPDCs derived from tibia with craniofacial hPDCs, from maxilla and mandible, respectively, each representing a potential source for cell-based tissue engineered implants for craniofacial applications. Maxilla and mandible-derived hPDCs display similar growth curves as tibial hPDCs, with equal trilineage differentiation potential toward chondrogenic, osteogenic, and adipogenic cells. These craniofacial hPDCs are positive for SSPC-markers CD73, CD164, and Podoplanin (PDPN), and negative for CD146, hematopoietic and endothelial lineage markers. Bulk RNA-sequencing identified genes that are differentially expressed between the three sources of hPDC. In particular, differential expression was found for genes of the HOX and DLX family, for SOX9 and genes involved in skeletal system development. The in vivo bone formation, 8 weeks after ectopic implantation in nude mice, was observed in constructs seeded with tibial and mandibular hPDCs. Taken together, we provide evidence that hPDCs show different profiles and properties according to their anatomical origin, and that craniofacial hPDCs are potential sources for cell-based bone tissue engineering strategies. The mandible-derived hPDCs display - both in vitro and in vivo - chondrogenic and osteogenic differentiation potential, which supports their future testing for use in craniofacial bone regeneration applications.
Collapse
Affiliation(s)
- Lisanne C Groeneveldt
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,OMFS IMPATH Research Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Tim Herpelinck
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Marina Maréchal
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Constantinus Politis
- OMFS IMPATH Research Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands.,Center for Biomics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA-R In Silico Medicine, Université de Liége, Liège, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Eskeatnaf Mulugeta
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Frank P Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
76
|
Gallinetti S, Burenstam Linder LK, Åberg J, Illies C, Engqvist H, Birgersson U. Titanium reinforced calcium phosphate improves bone formation and osteointegration in ovine calvaria defects: a comparative 52-weeks study. Biomed Mater 2020; 16. [PMID: 33181501 DOI: 10.1088/1748-605x/abca12] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 11/12/2020] [Indexed: 11/12/2022]
Abstract
In a 52-week ovine calvaria implantation model, the restoration of cranial defects with a bare titanium mesh (Ti-mesh) and a titanium mesh embedded in a calcium phosphate (CaP-Ti) were evaluated in seven animals. During the study, no major clinical abnormalities were observed, and all sheep presented a normal neurologic assessment. Blood and CSF analysis, made at termination, did not show any abnormalities. No indentation of the soft tissue was observed for either test article; however, the Ti-mesh burr-hole covers were associated with filling of the calvarial defect by fibrous tissue mainly. Some bone formation was observed at the bottom of the created defect, but no significant bone was formed in the proximity of the implant. The defect sites implanted with CaP-Ti were characterized by a moderate degradation of the calcium phosphate that was replaced by mature bone tissue. Calcium-phosphate-filled macrophages were observed in all animals, indicating that they might play a vital role in osteogenesis. The newly formed bone was present, especially at the bony edges of the defect and on the dura side. Integration of the titanium mesh in a calcium phosphate improved bone formation and osteointegration in comparison to a bare titanium mesh.
Collapse
Affiliation(s)
| | - Lars Kihlstrom Burenstam Linder
- Neurosurgery, Clinical Neuroscience Research Centre, Karolinska University Hospital, Eugeniav 3 Solna, Dartford, DA1 2EN, SWEDEN
| | - Jonas Åberg
- Department of Engineering Sciences, Applied Materials Science Section, Uppsala University, Uppsala, SWEDEN
| | - Christopher Illies
- Department of Clinical Pathology, Karolinska University Hospital, Stockholm, SWEDEN
| | - Håkan Engqvist
- Department of Engineering Sciences, Applied Materials Science Section, Uppsala Universitet, Uppsala, SWEDEN
| | - Ulrik Birgersson
- Clintec, Karolinska institutet Department of Clinical Sciences Intervention and Technology, Huddinge, SWEDEN
| |
Collapse
|
77
|
Impact of scaffold granule size use in Masquelet technique on periosteal reaction: a study in rat femur critical size bone defect model. Eur J Trauma Emerg Surg 2020; 48:679-687. [PMID: 33025170 PMCID: PMC8825401 DOI: 10.1007/s00068-020-01516-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Purpose The Masquelet technique for the treatment of large bone defects is a two-stage procedure based on an induced membrane. Compared to mature periosteum, the induced membrane differs significantly. However, both play a crucial role in bone regeneration. As part of a histological and radiological post-evaluation of an earlier project, we analyzed the influence of the granule size of the bone void filler Herafill® on development of periosteum regrowth in a critical size defect. Methods We compared three different sizes of Herafill® granules (Heraeus Medical GmbH, Wehrheim) in vivo in a rat femoral critical size defect (10 mm) treated with the induced membrane technique. After 8 weeks healing time, femurs were harvested and taken for histological and radiological analysis. Results A significantly increased regrowth of periosteum into the defect was found when small granules were used. Large granules showed significantly increased occurrence of bone capping. Small granules lead to significant increase in callus formation in the vicinity to the membrane. Conclusion The size of Herafill® granules has significant impact on the development of periosteal-like structures around the defect using Masquelet’s induced membrane technique. Small granules show significantly increased regrowth of periosteum and improved bone formation adjacent to the induced membrane.
Collapse
|
78
|
Xia C, Ge Q, Fang L, Yu H, Zou Z, Zhang P, Lv S, Tong P, Xiao L, Chen D, Wang PE, Jin H. TGF-β/Smad2 signalling regulates enchondral bone formation of Gli1 + periosteal cells during fracture healing. Cell Prolif 2020; 53:e12904. [PMID: 32997394 PMCID: PMC7653269 DOI: 10.1111/cpr.12904] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/20/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Most bone fracture heals through enchondral bone formation that relies on the involvement of periosteal progenitor cells. However, the identity of periosteal progenitor cells and the regulatory mechanism of their proliferation and differentiation remain unclear. The aim of this study was to investigate whether Gli1-CreERT2 can identify a population of murine periosteal progenitor cells and the role of TGF-β signalling in periosteal progenitor cells on fracture healing. MATERIALS AND METHODS Double heterozygous Gli1-CreERT2 ;Rosa26-tdTomatoflox/wt mice were sacrificed at different time points for tracing the fate of Gli1+ cells in both intact and fracture bone. Gli1-CreERT2 -mediated Tgfbr2 knockout (Gli1-CreERT2 ;Tgfbr2flox/flox ) mice were subjected to fracture surgery. At 4, 7, 10, 14 and 21 days post-surgery, tibia samples were harvested for tissue analyses including μCT, histology, real-time PCR and immunofluorescence staining. RESULTS Through cell lineage-tracing experiments, we have revealed that Gli1-CreER T2 can be used to identify a subpopulation of periosteal progenitor cells in vivo that persistently reside in periosteum and contribute to osteochondral elements during fracture repair. During the healing process, TGF-β signalling is continually activated in the reparative Gli1+ periosteal cells. Conditional knockout of Tgfbr2 in these cells leads to a delayed and impaired enchondral bone formation, at least partially due to the reduced proliferation and chondrogenic and osteogenic differentiation of Gli1+ periosteal cells. CONCLUSIONS TGF-β signalling plays an essential role on fracture repair via regulating enchondral bone formation process of Gli1+ periosteal cells.
Collapse
Affiliation(s)
- Chenjie Xia
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopedic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Qinwen Ge
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liang Fang
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huan Yu
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhen Zou
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peng Zhang
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuaijie Lv
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Luwei Xiao
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ping-Er Wang
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongting Jin
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
79
|
Ortinau LC, Wang H, Lei K, Deveza L, Jeong Y, Hara Y, Grafe I, Rosenfeld SB, Lee D, Lee B, Scadden DT, Park D. Identification of Functionally Distinct Mx1+αSMA+ Periosteal Skeletal Stem Cells. Cell Stem Cell 2020; 25:784-796.e5. [PMID: 31809737 DOI: 10.1016/j.stem.2019.11.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/11/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022]
Abstract
The periosteum is critical for bone maintenance and healing. However, the in vivo identity and specific regulatory mechanisms of adult periosteum-resident skeletal stem cells are unknown. Here, we report animal models that selectively and durably label postnatal Mx1+αSMA+ periosteal stem cells (P-SSCs) and establish that P-SSCs are a long-term repopulating, functionally distinct SSC subset responsible for lifelong generation of periosteal osteoblasts. P-SSCs rapidly migrate toward an injury site, supply osteoblasts and chondrocytes, and recover new periosteum. Notably, P-SSCs specifically express CCL5 receptors, CCR3 and CCR5. Real-time intravital imaging revealed that the treatment with CCL5 induces P-SSC migration in vivo and bone healing, while CCL5/CCR5 deletion, CCR5 inhibition, or local P-SSC ablation reduces osteoblast number and delays bone healing. Human periosteal cells express CCR5 and undergo CCL5-mediated migration. Thus, the adult periosteum maintains genetically distinct SSC subsets with a CCL5-dependent migratory mechanism required for bone maintenance and injury repair.
Collapse
Affiliation(s)
- Laura C Ortinau
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hamilton Wang
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Kevin Lei
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Lorenzo Deveza
- Department of Orthopedic Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Youngjae Jeong
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yannis Hara
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ingo Grafe
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Scott B Rosenfeld
- Texas Children's Hospital, 6701 Fannin Street, Houston, TX 77030, USA
| | - Dongjun Lee
- Department of Convergence of Medical Science, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Brendan Lee
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - David T Scadden
- Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Dongsu Park
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
80
|
Lammens J, Maréchal M, Delport H, Geris L, Oppermann H, Vukicevic S, Luyten FP. A cell-based combination product for the repair of large bone defects. Bone 2020; 138:115511. [PMID: 32599225 DOI: 10.1016/j.bone.2020.115511] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/14/2020] [Accepted: 06/14/2020] [Indexed: 01/19/2023]
Abstract
Regenerative cell-based implants using periosteum-derived stem cells were developed for the treatment of large 3 cm fresh and 4.5 centimeter biological compromised bone gaps in a tibial sheep model and compared with an acellular ceramic-collagen void filler. It was hypothesized that the latter is insufficient to heal large skeletal defects due to reduced endogenous biological potency. To this purpose a comparison was made between the ceramic dicalciumphosphate scaffold (CopiOs®) as such, the same ceramic coated with clinical grade Bone Morphogenetic Protein 2 and 6 (BMP) only or a BMP coated cell-seeded combination product. These implants were evaluated in 2 sheep models, a fresh 3 cm critical size tibial defect and a 4.5 cm biologically exhausted tibial defect. For the groups in which growth factors were applied, BMP-6 was chosen at a dose of 344 μg for 3 cm and 1.500 μg or 3.800 μg for 4.5 cm defects. An additional group in the 4.5 cm defect was tested using BMP-2 in a dose of 1.500 μg. For all the cell based implants autologous periosteum-derived cells were used which were cultured in monolayer during 6 weeks. For the fresh defect 408 million cells and for the biologically exhausted tibial defect 612 million cells were drop-seeded on the BMP coated scaffolds. Bone healing was studied during 16 weeks postimplantation, using standard radiographs. While fresh defects responded to all treatments, regardless the use of cells, the biologically hampered defects responded in half of the cases and only if the BMP-cell combination product was used, supporting the concept that cell-based therapies may become attractive in treating defects with a compromised biological status.
Collapse
Affiliation(s)
- Johan Lammens
- Department of Orthopaedic Surgery, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering of the KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - Marina Maréchal
- Prometheus, Division of Skeletal Tissue Engineering of the KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Skeletal Biology and Engineering Research Center, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Hendrik Delport
- Prometheus, Division of Skeletal Tissue Engineering of the KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering of the KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Skeletal Biology and Engineering Research Center, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300, 3001 Heverlee (Leuven), Belgium; Biomechanics Research Unit, GIGA In silico medicine, University of Liège, Quartier Hôpital, Avenue de l'Hôpital 1, 4000 Liège 1, Belgium
| | - Hermann Oppermann
- Genera Research, Svetonedeljska cesta 2, 10436 Kalinovica, Sveta Nedelja, Croatia
| | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Šalata ul. 2, 10000 Zagreb, Croatia
| | - Frank P Luyten
- Prometheus, Division of Skeletal Tissue Engineering of the KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Skeletal Biology and Engineering Research Center, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
81
|
Yu Y, Wang Y, Zhang W, Wang H, Li J, Pan L, Han F, Li B. Biomimetic periosteum-bone substitute composed of preosteoblast-derived matrix and hydrogel for large segmental bone defect repair. Acta Biomater 2020; 113:317-327. [PMID: 32574859 DOI: 10.1016/j.actbio.2020.06.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022]
Abstract
Repairing large segmental bone defects above a critical size remains challenging with high risk of delayed union or even non-union. From the perspective of bone development and clinical experience, periosteum plays an indispensable role in bone repair and reconstruction. In this study, we explored the feasibility of using preosteoblast-derived matrix (pODM) as a biomimetic periosteum. By culturing MC3T3-E1 cell sheet on poly(dimethylsiloxane) and performing decellularization, an integral cell-free sheet of pODM could be readily harvested. Bone marrow mesenchymal stem cells (BMSCs) adhered and proliferated well on pODM. In addition, pODM exhibited a chemotactic effect on BMSCs in a concentration-dependent manner and also promoted osteogenic differentiation of BMSCs. Following that, pODM was wrapped around a gelatin methacryloyl (GelMA) hydrogel to construct an engineered periosteum-bone substitute. A rabbit radius segmental bone defect model was used to examine the bone repair efficacy of pODM/GelMA. Upon implantation of pODM/GelMA construct for 12 weeks, the critical-sized bone defects completely healed with remarkable full reconstruction of medullary cavity at the radial diaphysis. Together, this work proposes a high potency of using precursor cell-derived matrix as a biomimetic periosteum, which preserves the beneficial biological factors while avoids the limitations of using exogenous cells for bone regeneration. Combining precursor cell-derived matrix with hydrogel may provide a promising periosteum-bone biomimetic substitute for bone repair. STATEMENT OF SIGNIFICANCE: Repairing large segmental bone defects above a critical size remains challenging. As the periosteum plays an essential role in bone repair, this study aimed to explore the use of preosteoblast-derived matrix (pODM), harvested from decellularized MC3T3-E1 cell sheet, as a biomimetic periosteum to facilitate bone repair. We found that in vitro, pODM exhibited considerable chemotactic effect and osteogenic induction capability to bone marrow mesenchymal stem cells (BMSCs). In vivo, implantation of pODM/gelatin methacryloyl (GelMA) constructs as engineered periosteum-bone substitutes effectively repaired the critical-sized segmental bone defects at rabbit radius. Surprisingly, remarkable full reconstruction of medullary cavity at the diaphysis was achieved. Therefore, combining pODM with hydrogel may provide a promising biomimetic substitute for bone repair.
Collapse
|
82
|
Li H, Wang H, Pan J, Li J, Zhang K, Duan W, Liang H, Chen K, Geng D, Shi Q, Yang H, Li B, Chen H. Nanoscaled Bionic Periosteum Orchestrating the Osteogenic Microenvironment for Sequential Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:36823-36836. [PMID: 32706234 DOI: 10.1021/acsami.0c06906] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Periosteum orchestrates bone repair. Previously developed artificial periosteum was mainly focusing on materials modification to simply enhance bone formation, but few were attempting to make the artificial periosteum fit different bone repair stages. Here, we constructed a functionalized periosteum, which was composed of an electrospun scaffold grafted with leptin receptor antibody (LepR-a) and BMP2-loaded hollow MnO2 (h-MnO2) nanoparticles through a polydopamine (PDA)-assisted technique. The bionic periosteum showed suitable mechanical properties and favorable biocompatibility. It effectively recruited skeletal stem cells (SSCs) through antigen-antibody interactions, as in in vitro cell adhesion tests, we observed that more SSCs attached to the LepR-a-grafted periosteum compared to the control group. In vivo, the LepR-a-grafted periosteum covered on the cranial defect in Prx1-Cre/ERT2, -EGFP mice recruited more Prx1-EGFP cells to the fracture site compared to control groups at post-surgery day 3, 7, and 14. Co-staining with Sp7 indicated that most of the recruited Prx1-EGFP cells underwent osteogenic lineage commitment. Sustained BMP2 release from h-MnO2 promoted osteogenesis by accelerating the osteogenic differentiation of recruited SSCs, as demonstrated by alkaline phosphatase (ALP) and alizarin red staining (ARS) in vitro and microcomputed tomography (micro-CT) in vivo. Interestingly, we also observed the growth of osteogenic coupled capillaries (CD31hiEmcnhi) in the bone repair site, which might be induced by increased platelet-derived growth factor-BB (PDGF-BB) in the regenerative microenvironment subsequent to SSCs' differentiation. Taken together, the findings from this study indicate that the multifunctionalized periosteum efficiently recruited and motivated the SSCs in vivo and orchestrated the osteogenic microenvironment for bone repair in a sequence manner. Thus, the construction of the bionic periosteum to couple with natural bone regeneration stages has been demonstrated to be effective in facilitating bone healing.
Collapse
Affiliation(s)
- Hanwen Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Huan Wang
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Jun Pan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Jiaying Li
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Kai Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Weifeng Duan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Huan Liang
- Medical College, Yangzhou University, 136 Jiangyang Road, Yangzhou, Jiangsu 225009, P. R. China
| | - Kangwu Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Dechun Geng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
| | - Qin Shi
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Bin Li
- Orthopedic Institute, Medical College, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215000, P. R. China
| | - Hao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215000, P. R. China
- Medical College, Yangzhou University, 136 Jiangyang Road, Yangzhou, Jiangsu 225009, P. R. China
| |
Collapse
|
83
|
Hellwinkel JE, Miclau T, Provencher MT, Bahney CS, Working ZM. The Life of a Fracture: Biologic Progression, Healing Gone Awry, and Evaluation of Union. JBJS Rev 2020; 8:e1900221. [PMID: 32796195 PMCID: PMC11147169 DOI: 10.2106/jbjs.rvw.19.00221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
New knowledge about the molecular biology of fracture-healing provides opportunities for intervention and reduction of risk for specific phases that are affected by disease and medications. Modifiable and nonmodifiable risk factors can prolong healing, and the informed clinician should optimize each patient to provide the best chance for union. Techniques to monitor progression of fracture-healing have not changed substantially over time; new objective modalities are needed.
Collapse
Affiliation(s)
- Justin E Hellwinkel
- Department of Orthopedic Surgery, New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Theodore Miclau
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Matthew T Provencher
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Chelsea S Bahney
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Zachary M Working
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
- Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
84
|
Kram V, Shainer R, Jani P, Meester JAN, Loeys B, Young MF. Biglycan in the Skeleton. J Histochem Cytochem 2020; 68:747-762. [PMID: 32623936 DOI: 10.1369/0022155420937371] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Small leucine rich proteoglycans (SLRPs), including Biglycan, have key roles in many organ and tissue systems. The goal of this article is to review the function of Biglycan and other related SLRPs in mineralizing tissues of the skeleton. The review is divided into sections that include Biglycan's role in structural biology, signaling, craniofacial and long bone homeostasis, remodeled skeletal tissues, and in human genetics. While many cell types in the skeleton are now known to be affected by Biglycan, there are still unanswered questions about its mechanism of action(s).
Collapse
Affiliation(s)
- Vardit Kram
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| | - Reut Shainer
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| | - Priyam Jani
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| | - Josephina A N Meester
- Laboratory of Cardiogenetics, Center of Medical Genetics, University Hospital Antwerp, University of Antwerp, Antwerp, Belgium
| | - Bart Loeys
- Laboratory of Cardiogenetics, Center of Medical Genetics, University Hospital Antwerp, University of Antwerp, Antwerp, Belgium
| | - Marian F Young
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| |
Collapse
|
85
|
Al Hosni R, Shah M, Cheema U, Roberts HC, Luyten FP, Roberts SJ. Mapping human serum-induced gene networks as a basis for the creation of biomimetic periosteum for bone repair. Cytotherapy 2020; 22:424-435. [PMID: 32522398 DOI: 10.1016/j.jcyt.2020.03.434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/21/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The periosteum is a highly vascularized, collagen-rich tissue that plays a crucial role in directing bone repair. This is orchestrated primarily by its resident progenitor cell population. Indeed, preservation of periosteum integrity is critical for bone healing. Cells extracted from the periosteum retain their osteochondrogenic properties and as such are a promising basis for tissue engineering strategies for the repair of bone defects. However, the culture expansion conditions and the way in which the cells are reintroduced to the defect site are critical aspects of successful translation. Indeed, expansion in human serum and implantation on biomimetic materials has previously been shown to improve in vivo bone formation. AIM This study aimed to develop a protocol to allow for the expansion of human periosteum derived cells (hPDCs) in a biomimetic periosteal-like environment. METHODS The expansion conditions were defined through the investigation of the bioactive cues involved in augmenting hPDC proliferative and multipotency characteristics, based on transcriptomic analysis of cells cultured in human serum. RESULTS Master regulators of transcriptional networks were identified, and an optimized periosteum-derived growth factor cocktail (PD-GFC; containing β-estradiol, FGF2, TNFα, TGFβ, IGF-1 and PDGF-BB) was generated. Expansion of hPDCs in PD-GFC resulted in serum mimicry with regard to the cell morphology, proliferative capacity and chondrogenic differentiation. When incorporated into a three-dimensional collagen type 1 matrix and cultured in PD-GFC, the hPDCs migrated to the surface that represented the matrix topography of the periosteum cambium layer. Furthermore, gene expression analysis revealed a down-regulated WNT and TGFβ signature and an up-regulation of CREB, which may indicate the hPDCs are recreating their progenitor cell signature. CONCLUSION This study highlights the first stage in the development of a biomimetic periosteum, which may have applications in bone repair.
Collapse
Affiliation(s)
- Rawiya Al Hosni
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Mittal Shah
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Umber Cheema
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Helen C Roberts
- Department of Natural Sciences, Faculty of Science & Technology, Middlesex University, London, UK
| | - Frank P Luyten
- Skeletal Biology and Tissue Engineering Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium and
| | - Scott J Roberts
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK; Skeletal Biology and Tissue Engineering Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium and; Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK.
| |
Collapse
|
86
|
Three-Dimensional-Printed Poly-L-Lactic Acid Scaffolds with Different Pore Sizes Influence Periosteal Distraction Osteogenesis of a Rabbit Skull. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7381391. [PMID: 32382570 PMCID: PMC7196141 DOI: 10.1155/2020/7381391] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/29/2020] [Accepted: 03/17/2020] [Indexed: 12/25/2022]
Abstract
The repair of bone defects is a big challenge in reconstructive surgery. Periosteal distraction osteogenesis (PDO), as a promising technique used for bone regeneration, forms a space between the periosteum and bone cortex to regenerate the new bone merely by distracting the periosteum. In order to investigate the influence of distractor framework on the PDO, we utilized three-dimensional (3D) printing technology to fabricate three kinds of poly-L-lactic acid (PLLA) scaffolds with different pore sizes in this study. The in vitro experiments showed that the customized PLLA scaffolds had different-sized microchannels with low toxicity, good biocompatibility, and enough mechanical strength. Then, we built up an in vivo bioreactor under the skull periosteum of New Zealand white rabbits. The distractors with different pore sizes all could satisfy the demand of periosteal distraction in the animal experiments. After 8 weeks of consolidation period, the quality and quantity of the newly formed bone were improved with the increasing pore sizes of the distractors. Moreover, the newly formed bone also displayed an increasing degree of vascularization. In conclusion, 3D printing technology could promote the innovation of PDO devices and fabricate optimized scaffolds with appropriate pore sizes, shapes, and structures. It would help us regenerate more functional tissue-engineered bone and provide new ideas for further clinical application of the PDO technique.
Collapse
|
87
|
Serowoky MA, Arata CE, Crump JG, Mariani FV. Skeletal stem cells: insights into maintaining and regenerating the skeleton. Development 2020; 147:147/5/dev179325. [PMID: 32161063 DOI: 10.1242/dev.179325] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Skeletal stem cells (SSCs) generate the progenitors needed for growth, maintenance and repair of the skeleton. Historically, SSCs have been defined as bone marrow-derived cells with inconsistent characteristics. However, recent in vivo tracking experiments have revealed the presence of SSCs not only within the bone marrow but also within the periosteum and growth plate reserve zone. These studies show that SSCs are highly heterogeneous with regard to lineage potential. It has also been revealed that, during digit tip regeneration and in some non-mammalian vertebrates, the dedifferentiation of osteoblasts may contribute to skeletal regeneration. Here, we examine how these research findings have furthered our understanding of the diversity and plasticity of SSCs that mediate skeletal maintenance and repair.
Collapse
Affiliation(s)
- Maxwell A Serowoky
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Claire E Arata
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Francesca V Mariani
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
88
|
Lipid availability determines fate of skeletal progenitor cells via SOX9. Nature 2020; 579:111-117. [PMID: 32103177 PMCID: PMC7060079 DOI: 10.1038/s41586-020-2050-1] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
The avascular nature of cartilage makes it a unique tissue1–4, but whether and how the absence of nutrient supply regulates chondrogenesis remains unknown. Here, we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate FoxO transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing fatty acid oxidation, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FoxOs during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.
Collapse
|
89
|
Batista JM, Nakagaki WR, Soares EA, Camilli JA. Effects of low-intensity pulsed ultrasound exposure on rats tibia periosteum. AN ACAD BRAS CIENC 2020; 92:e20180903. [PMID: 32074178 DOI: 10.1590/0001-3765202020180903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/10/2018] [Indexed: 12/15/2022] Open
Abstract
The periosteum is a rich source of osteoprogenitor cells and periosteal grafts can be used as an alternative method to replace bone grafts. The low-intensity pulsed ultrasound (LIPUS) has often been used as a noninvasive method to stimulate osteogenesis and reduce the fracture healing time. The aim of this study was to evaluate the effects of the ultrasound exposure on the rat tibia periosteum. Group I (7 animals) received LIPUS therapy on the left tibia for 7 days and group II (7 animals) on the left tibia for 14 days. After euthanasia, the tibias were processed. Number of periosteal cells and vessels and thickness of the periosteum were analyzed. The number of periosteal cells was higher in stimulated periosteum compared to controls at 7 and 14 days, but the number of vessels and the thickness only were higher in the group stimulated at 14 days. Furthermore, the ultrasound treatment for 14 days was more effective than 7 days. The ultrasound stimulation of the periosteum prior to grafting procedure can be advantageous, since it increases periosteal activity, and LIPUS may be an alternative method for stimulating the periosteum when the use of periosteal grafts in bone repair is needed.
Collapse
Affiliation(s)
- Jaqueline Martins Batista
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas/UNICAMP, Programa de Pós-Graduação em Biologia Celular e Estrutural, Avenida Bertrand Russel, s/n, 13083-865 Campinas, SP, Brazil
| | - Wilson Romero Nakagaki
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas/UNICAMP, Programa de Pós-Graduação em Biologia Celular e Estrutural, Avenida Bertrand Russel, s/n, 13083-865 Campinas, SP, Brazil.,Programa de Mestrado em Ciências da Saúde, Universidade do Oeste Paulista/UNOESTE, Pró-Reitoria de Pesquisa e Pós-Graduação/Campus II, Rodovia Raposo Tavares, Km 572, Bairro do Limoeiro, 19067-175 Presidente Prudente, SP, Brazil
| | - Evelise Aline Soares
- Departamento de Anatomia, Universidade Federal de Alfenas/UNIFAL, Rua Gabriel Monteiro da Silva 700, 37130-001 Alfenas, MG, Brazil
| | - José Angelo Camilli
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas/UNICAMP, Programa de Pós-Graduação em Biologia Celular e Estrutural, Avenida Bertrand Russel, s/n, 13083-865 Campinas, SP, Brazil
| |
Collapse
|
90
|
Umrath F, Weber M, Reinert S, Wendel HP, Avci-Adali M, Alexander D. iPSC-Derived MSCs Versus Originating Jaw Periosteal Cells: Comparison of Resulting Phenotype and Stem Cell Potential. Int J Mol Sci 2020; 21:ijms21020587. [PMID: 31963278 PMCID: PMC7013802 DOI: 10.3390/ijms21020587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/04/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cell-derived mesenchymal stem cell-like cells (iMSCs) are considered to be a promising source of progenitor cells for approaches in the field of bone regeneration. In a previous study, we described the generation of footprint-free induced pluripotent stem cells (iPSCs) from human jaw periosteal cells (JPCs) by transfection of a self-replicating RNA (srRNA) and subsequent differentiation into functional osteogenic progenitor cells. In order to facilitate the prospective transfer into clinical practice, xeno-free reprogramming and differentiation methods were established. In this study, we compared the properties and stem cell potential of the iMSCs produced from JPC-derived iPSCs with the parental primary JPCs they were generated from. Our results demonstrated, on the one hand, a comparable differentiation potential of iMSCs and JPCs. Additionally, iMSCs showed significantly longer telomere lengths compared to JPCs indicating rejuvenation of the cells during reprogramming. On the other hand, proliferation, mitochondrial activity, and senescence-associated beta-galactosidase (SA-β-gal) activity indicated early senescence of iMSCs. These data demonstrate the requirement of further optimization strategies to improve mesenchymal development of JPC-derived iPSCs in order to take advantage of the best features of reprogrammed and rejuvenated cells.
Collapse
Affiliation(s)
- Felix Umrath
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.U.); (S.R.)
| | - Marbod Weber
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (M.W.); (H.-P.W.); (M.A.-A.)
| | - Siegmar Reinert
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.U.); (S.R.)
| | - Hans-Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (M.W.); (H.-P.W.); (M.A.-A.)
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (M.W.); (H.-P.W.); (M.A.-A.)
| | - Dorothea Alexander
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany; (F.U.); (S.R.)
- Correspondence: ; Tel.: +49-7071-29-82418
| |
Collapse
|
91
|
Qiu P, Li M, Chen K, Fang B, Chen P, Tang Z, Lin X, Fan S. Periosteal matrix-derived hydrogel promotes bone repair through an early immune regulation coupled with enhanced angio- and osteogenesis. Biomaterials 2020; 227:119552. [DOI: 10.1016/j.biomaterials.2019.119552] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 01/15/2023]
|
92
|
Wang D, Gilbert JR, Zhang X, Zhao B, Ker DFE, Cooper GM. Calvarial Versus Long Bone: Implications for Tailoring Skeletal Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2019; 26:46-63. [PMID: 31588853 DOI: 10.1089/ten.teb.2018.0353] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue-engineered graft substitutes have shown great potential to treat large bone defects. While we usually assume that therapeutic approaches developed for appendicular bone healing could be similarly translated for application in craniofacial reconstruction and vice versa, this is not necessarily accurate. In addition to those more well-known healing-associated factors, such as age, lifestyle (e.g., nutrition and smoking), preexisting disease (e.g., diabetes), medication, and poor blood supply, the developmental origins and surrounding tissue of the wound sites can largely affect the fracture healing outcome as well as designed treatments. Therefore, the strategies developed for long bone fracture repair might not be suitable or directly applicable to skull bone repair. In this review, we discuss aspects of development, healing process, structure, and tissue engineering considerations between calvarial and long bones to assist in designing the tailored bone repair strategies. Impact Statement We summarized, in this review, the existing body of knowledge between long bone and calvarial bone with regard to their development and healing, tissue structure, and consideration of current tissue engineering strategies. By highlighting their similarities and differences, we propose that tailored tissue engineering strategies, such as scaffold features, growth factor usage, and the source of cells for tissue or region-specific bone repair, are necessary to ensure an optimized healing outcome.
Collapse
Affiliation(s)
- Dan Wang
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James R Gilbert
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingkun Zhao
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gregory M Cooper
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
93
|
Xu T, Yu X, Yang Q, Liu X, Fang J, Dai X. Autologous Micro-Fragmented Adipose Tissue as Stem Cell-Based Natural Scaffold for Cartilage Defect Repair. Cell Transplant 2019; 28:1709-1720. [PMID: 31565996 PMCID: PMC6923561 DOI: 10.1177/0963689719880527] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Osteoarthritis (OA) poses a tough challenge worldwide. Adipose-derived stem cells (ASCs)
have been proved to play a promising role in cartilage repair. However, enzymatic
digestion, ex vivo culture and expansion, with significant senescence and decline in
multipotency, limit their application. The present study was designed to obtain
micro-fragmented adipose tissue (MFAT) through gentle mechanical force and determine the
effect of this stem cell-based natural scaffold on repair of full-thickness cartilage
defects. In this study, ASCs sprouted from MFAT were characterized by
multi-differentiation induction and flow cytometry. Scratch and transwell migration assays
were operated to determine whether MFAT could promote migration of chondrocytes in vitro.
In a rat model, cartilage defects were created on the femoral groove and treated with
intra-articular injection of MFAT or PBS for 6 weeks and 12 weeks (n =
12). At the time points, the degree of cartilage repair was evaluated by histological
staining, immunohistochemistry and scoring, respectively. Two unoperated age-matched
animals served as native controls. ASCs derived from MFAT possessed properties to
differentiate into adipocytes, osteocytes and chondrocytes, with expression of mesenchymal
stem cell markers (CD29, 44, 90) and no expression of hematopoietic markers (CD31, 34,
45). In addition, MFAT could significantly promote migration of chondrocytes. MFAT-treated
defects showed improved macroscopic appearance and histological evaluation compared with
PBS-treated defects at both time points. After 12 weeks of treatment, MFAT-treated defects
displayed regular surface, high amount of hyaline cartilage, intact subchondral bone
reconstruction and corresponding formation of type I, II, and VI collagen, which resembled
the normal cartilage. This study demonstrates the efficacy of MFAT on cartilage repair in
an animal model for the first time, and the utility of MFAT as a ready-to-use therapeutic
alternative to traditional stem cell therapy.
Collapse
Affiliation(s)
- Tengjing Xu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinning Yu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Orthopaedic Surgery, Hangzhou Mingzhou Hospital (International Medical Center, Second Affiliated Hospital, Zhejiang University School of Medicine), Hangzhou, China
| | - Quanming Yang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaonan Liu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghua Fang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Orthopaedic Surgery, Hangzhou Mingzhou Hospital (International Medical Center, Second Affiliated Hospital, Zhejiang University School of Medicine), Hangzhou, China
| | - Xuesong Dai
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Orthopaedic Surgery, Hangzhou Mingzhou Hospital (International Medical Center, Second Affiliated Hospital, Zhejiang University School of Medicine), Hangzhou, China
| |
Collapse
|
94
|
Bone growth as the main determinant of mouse digit tip regeneration after amputation. Sci Rep 2019; 9:9720. [PMID: 31273239 PMCID: PMC6609708 DOI: 10.1038/s41598-019-45521-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/08/2019] [Indexed: 01/08/2023] Open
Abstract
Regeneration is classically demonstrated in mammals using mice digit tip. In this study, we compared different amputation plans and show that distally amputated digits regrow with morphology close to normal but fail to regrow the fat pad. Proximally amputated digits do not regrow the phalangeal bone, but the remaining structures (nail, skin and connective tissue), all with intrinsic regenerative capacity, re-establishing integrity indistinguishably in distally and proximally amputated digits. Thus, we suggest that the bone growth promoted by signals and progenitor cells not removed by distal amputations is responsible for the re-establishment of a drastically different final morphology after distal or proximal digit tip amputations. Despite challenging the use of mouse digit tip as a model system for limb regeneration in mammals, these findings evidence a main role of bone growth in digit tip regeneration and suggest that mechanisms that promote joint structures formation should be the main goal of regenerative medicine for limb and digit regrowth.
Collapse
|
95
|
Brekelmans C, Hollants S, De Groote C, Sohier N, Maréchal M, Geris L, Luyten FP, Ginckels L, Sciot R, de Ravel T, De Smet L, Lammens J, Legius E, Brems H. Neurofibromatosis type 1-related pseudarthrosis: Beyond the pseudarthrosis site. Hum Mutat 2019; 40:1760-1767. [PMID: 31066482 DOI: 10.1002/humu.23783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 05/03/2019] [Accepted: 05/04/2019] [Indexed: 12/23/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder affecting approximately 1 in 2,000 newborns. Up to 5% of NF1 patients suffer from pseudarthrosis of a long bone (NF1-PA). Current treatments are often unsatisfactory, potentially leading to amputation. To gain more insight into the pathogenesis we cultured cells from PA tissue and normal-appearing periosteum of the affected bone for NF1 mutation analysis. PA cells were available from 13 individuals with NF1. Biallelic NF1 inactivation was identified in all investigated PA cells obtained during the first surgery. Three of five cases sampled during a later intervention showed biallelic NF1 inactivation. Also, in three individuals, we examined periosteum-derived cells from normal-appearing periosteum proximal and distal to the PA. We identified the same biallelic NF1 inactivation in the periosteal cells outside the PA region. These results indicate that NF1 inactivation is required but not sufficient for the development of NF1-PA. We observed that late-onset NF1-PA occurs and is not always preceded by congenital bowing. Furthermore, the failure to identify biallelic inactivation in two of five later interventions and one reintervention with a known somatic mutation indicates that NF1-PA can persist after the removal of most NF1 negative cells.
Collapse
Affiliation(s)
- Carlijn Brekelmans
- Department of Human Genetics, KU Leuven-University of Leuven, Leuven, Belgium
| | - Silke Hollants
- Clinical Department of Human Genetics, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Caroline De Groote
- Clinical Department of Human Genetics, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Natalie Sohier
- Clinical Department of Human Genetics, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Marina Maréchal
- Department of Development and Regeneration, Prometheus LRD Division of Skeletal Tissue Engineering, KU Leuven-University of Leuven, Leuven, Belgium
| | - Liesbet Geris
- Department of Mechanical Engineering, Prometheus LRD Division of Skeletal Tissue Engineering, KU Leuven-University of Leuven, Leuven, Belgium.,GIGA In Silico Medicine, University of Liège, Liège, Belgium
| | - Frank P Luyten
- Department of Development and Regeneration, Prometheus LRD Division of Skeletal Tissue Engineering, KU Leuven-University of Leuven, Leuven, Belgium.,Department of Rheumatology, KU Leuven-University Hospitals Leuven, Leuven, Belgium
| | - Lieve Ginckels
- Department of Orthopaedic Surgery, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Raf Sciot
- Department of Imaging and Pathology, KU Leuven-University of Leuven, Leuven, Belgium.,Department of Pathology, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Thomy de Ravel
- Department of Human Genetics, KU Leuven-University of Leuven, Leuven, Belgium.,Clinical Department of Human Genetics, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Luc De Smet
- Department of Orthopaedic Surgery, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration, KU Leuven-University of Leuven, Leuven, Belgium
| | - Johan Lammens
- Department of Development and Regeneration, Prometheus LRD Division of Skeletal Tissue Engineering, KU Leuven-University of Leuven, Leuven, Belgium.,Department of Orthopaedic Surgery, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, KU Leuven-University of Leuven, Leuven, Belgium.,Clinical Department of Human Genetics, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Hilde Brems
- Department of Human Genetics, KU Leuven-University of Leuven, Leuven, Belgium.,Clinical Department of Human Genetics, KU Leuven-University of Leuven, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
96
|
Kudva AK, Dikina AD, Luyten FP, Alsberg E, Patterson J. Gelatin microspheres releasing transforming growth factor drive in vitro chondrogenesis of human periosteum derived cells in micromass culture. Acta Biomater 2019; 90:287-299. [PMID: 30905864 PMCID: PMC6597958 DOI: 10.1016/j.actbio.2019.03.039] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/14/2019] [Accepted: 03/20/2019] [Indexed: 11/29/2022]
Abstract
For cartilage tissue engineering, several in vitro culture methodologies have displayed potential for the chondrogenic differentiation of mesenchymal stem cells (MSCs). Micromasses, cell aggregates or pellets, and cell sheets are all structures with high cell density that provides for abundant cell-cell interactions, which have been demonstrated to be important for chondrogenesis. Recently, these culture systems have been improved via the incorporation of growth factor releasing components such as degradable microspheres within the structures, further enhancing chondrogenesis. Herein, we incorporated different amounts of gelatin microspheres releasing transforming growth factor β1 (TGF-β1) into micromasses composed of human periosteum derived cells (hPDCs), an MSC-like cell population. The aim of this research was to investigate chondrogenic stimulation by TGF-β1 delivery from these degradable microspheres in comparison to exogenous supplementation with TGF-β1 in the culture medium. Microscopy showed that the gelatin microspheres could be successfully incorporated within hPDC micromasses without interfering with the formation of the structure, while biochemical analysis and histology demonstrated increasing DNA content at week 2 and accumulation of glycosaminoglycan and collagen at weeks 2 and 4. Importantly, similar chondrogenesis was achieved when TGF-β1 was delivered from the microspheres compared to controls with TGF-β1 in the medium. Increasing the amount of growth factor within the micromasses by increasing the amount of microspheres added did not further improve chondrogenesis of the hPDCs. These findings demonstrate the potential of using cytokine releasing, gelatin microspheres to enhance the chondrogenic capabilities of hPDC micromasses as an alternative to supplementation of the culture medium with growth factors. STATEMENT OF SIGNIFICANCE: Gelatin microspheres are utilized for growth factor delivery to enhance chondrogenesis of mesenchymal stem cells (MSCs) in high cell density culture systems. Herein, we employ a new combination of these microspheres with micromasses of human periosteum-derived cells, which possess ease of isolation, excellent expansion potential, and MSC-like differentiation capabilities. The resulting localized delivery of transforming growth factor β1 increases glycosaminoglycan and collagen production within the micromasses without exogenous stimulation in the medium. This unique combination is able to drive chondrogenesis up to similar levels as seen in micromasses that do receive exogenous stimulation. The addition of growth factor releasing microspheres to high cell density micromasses has the potential to reduce costs associated with this strategy for cartilage tissue engineering.
Collapse
Affiliation(s)
- Abhijith K Kudva
- Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44, box 2450, 3001 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Herestraat 49, box 813, 3000 Leuven, Belgium; Skeletal Biology and Engineering Research Center, KU Leuven, Herestraat 49, box 813, 3000 Leuven, Belgium.
| | - Anna D Dikina
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Frank P Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Herestraat 49, box 813, 3000 Leuven, Belgium; Skeletal Biology and Engineering Research Center, KU Leuven, Herestraat 49, box 813, 3000 Leuven, Belgium.
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Jennifer Patterson
- Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44, box 2450, 3001 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Herestraat 49, box 813, 3000 Leuven, Belgium; Department of Imaging & Pathology, KU Leuven, Kapucijnenvoer 7 block a, box 7001, 3000 Leuven, Belgium.
| |
Collapse
|
97
|
Gupta P, Hall GN, Geris L, Luyten FP, Papantoniou I. Human Platelet Lysate Improves Bone Forming Potential of Human Progenitor Cells Expanded in Microcarrier-Based Dynamic Culture. Stem Cells Transl Med 2019; 8:810-821. [PMID: 31038850 PMCID: PMC6646698 DOI: 10.1002/sctm.18-0216] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/19/2019] [Indexed: 12/22/2022] Open
Abstract
Xenogeneic‐free media are required for translating advanced therapeutic medicinal products to the clinics. In addition, process efficiency is crucial for ensuring cost efficiency, especially when considering large‐scale production of mesenchymal stem cells (MSCs). Human platelet lysate (HPL) has been increasingly adopted as an alternative for fetal bovine serum (FBS) for MSCs. However, its therapeutic and regenerative potential in vivo is largely unexplored. Herein, we compare the effects of FBS and HPL supplementation for a scalable, microcarrier‐based dynamic expansion of human periosteum‐derived cells (hPDCs) while assessing their bone forming capacity by subcutaneous implantation in small animal model. We observed that HPL resulted in faster cell proliferation with a total fold increase of 5.2 ± 0.61 in comparison to 2.7 ± 02.22‐fold in FBS. Cell viability and trilineage differentiation capability were maintained by HPL, although a suppression of adipogenic differentiation potential was observed. Differences in mRNA expression profiles were also observed between the two on several markers. When implanted, we observed a significant difference between the bone forming capacity of cells expanded in FBS and HPL, with HPL supplementation resulting in almost three times more mineralized tissue within calcium phosphate scaffolds. FBS‐expanded cells resulted in a fibrous tissue structure, whereas HPL resulted in mineralized tissue formation, which can be classified as newly formed bone, verified by μCT and histological analysis. We also observed the presence of blood vessels in our explants. In conclusion, we suggest that replacing FBS with HPL in bioreactor‐based expansion of hPDCs is an optimal solution that increases expansion efficiency along with promoting bone forming capacity of these cells. stem cells translational medicine2019;8:810&821
Collapse
Affiliation(s)
- Priyanka Gupta
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Gabriella Nilsson Hall
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA-R In Silico Medicine, Université de Liege, Liège, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Frank P Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
98
|
Tam WL, Luyten FP, Roberts SJ. From skeletal development to the creation of pluripotent stem cell-derived bone-forming progenitors. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0218. [PMID: 29786553 DOI: 10.1098/rstb.2017.0218] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Bone has many functions. It is responsible for protecting the underlying soft organs, it allows locomotion, houses the bone marrow and stores minerals such as calcium and phosphate. Upon damage, bone tissue can efficiently repair itself. However, healing is hampered if the defect exceeds a critical size and/or is in compromised conditions. The isolation or generation of bone-forming progenitors has applicability to skeletal repair and may be used in tissue engineering approaches. Traditionally, bone engineering uses osteochondrogenic stem cells, which are combined with scaffold materials and growth factors. Despite promising preclinical data, limited translation towards the clinic has been observed to date. There may be several reasons for this including the lack of robust cell populations with favourable proliferative and differentiation capacities. However, perhaps the most pertinent reason is the failure to produce an implant that can replicate the developmental programme that is observed during skeletal repair. Pluripotent stem cells (PSCs) can potentially offer a solution for bone tissue engineering by providing unlimited cell sources at various stages of differentiation. In this review, we summarize key embryonic signalling pathways in bone formation coupled with PSC differentiation strategies for the derivation of bone-forming progenitors.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Wai Long Tam
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Frank P Luyten
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Scott J Roberts
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium .,Bone Therapeutic Area, UCB Pharma, 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| |
Collapse
|
99
|
Kuwahara ST, Serowoky MA, Vakhshori V, Tripuraneni N, Hegde NV, Lieberman JR, Crump JG, Mariani FV. Sox9+ messenger cells orchestrate large-scale skeletal regeneration in the mammalian rib. eLife 2019; 8:40715. [PMID: 30983567 PMCID: PMC6464605 DOI: 10.7554/elife.40715] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/22/2019] [Indexed: 11/13/2022] Open
Abstract
Most bones in mammals display a limited capacity for natural large-scale repair. The ribs are a notable exception, yet the source of their remarkable regenerative ability remains unknown. Here, we identify a Sox9-expressing periosteal subpopulation that orchestrates large-scale regeneration of murine rib bones. Deletion of the obligate Hedgehog co-receptor, Smoothened, in Sox9-expressing cells prior to injury results in a near-complete loss of callus formation and rib bone regeneration. In contrast to its role in development, Hedgehog signaling is dispensable for the proliferative expansion of callus cells in response to injury. Instead, Sox9-positive lineage cells require Hh signaling to stimulate neighboring cells to differentiate via an unknown signal into a skeletal cell type with dual chondrocyte/osteoblast properties. This type of callus cell may be critical for bridging large bone injuries. Thus despite contributing to only a subset of callus cells, Sox9-positive progenitors play a major role in orchestrating large-scale bone regeneration. Editorial note This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Stephanie T Kuwahara
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, United States
| | - Maxwell A Serowoky
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, United States
| | - Venus Vakhshori
- Department of Orthopaedic Surgery, University of Southern California, Keck School of Medicine, Los Angeles, United States
| | - Nikita Tripuraneni
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, United States
| | - Neel V Hegde
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, United States
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, University of Southern California, Keck School of Medicine, Los Angeles, United States
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, United States
| | - Francesca V Mariani
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Keck School of Medicine, Los Angeles, United States
| |
Collapse
|
100
|
Verbeeck L, Geris L, Tylzanowski P, Luyten FP. Uncoupling of in-vitro identity of embryonic limb derived skeletal progenitors and their in-vivo bone forming potential. Sci Rep 2019; 9:5782. [PMID: 30962493 PMCID: PMC6453955 DOI: 10.1038/s41598-019-42259-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/27/2019] [Indexed: 01/15/2023] Open
Abstract
The healing of large bone defects remains a major unmet medical need. Our developmental engineering approach consists of the in vitro manufacturing of a living cartilage tissue construct that upon implantation forms bone by recapitulating an endochondral ossification process. Key to this strategy is the identification of the cells to produce such cartilage intermediates efficiently. We applied a cell selection strategy based on published skeletal stem cell markers using mouse embryonic limb cartilage as cell source and analysed their potential to form bone in an in vivo ectopic assay. FGF2 supplementation to the culture media for expansion blocked dedifferentiation of the embryonic cartilage cells in culture and enriched for stem cells and progenitors as quantified using the recently published CD marker set. However, when the stem cells and progenitors were fractionated from expanded embryonic cartilage cells and assessed in the ectopic assay, a major loss of bone forming potential was observed. We conclude that cell expansion appears to affect the association between cell identity based on CD markers and in vivo bone forming capacity.
Collapse
Affiliation(s)
- Louca Verbeeck
- Prometheus, Div of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Tissue Engineering laboratory, SBERC, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Div of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, University of Liege, Liege, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Przemko Tylzanowski
- Development & Stem Cell Biology laboratory, SBERC, KU Leuven, Leuven, Belgium.,Dept of Bioch. & Mol Biol., Medical University Lublin, Lublin, Poland
| | - Frank P Luyten
- Prometheus, Div of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium. .,Tissue Engineering laboratory, SBERC, KU Leuven, Leuven, Belgium. .,Development & Stem Cell Biology laboratory, SBERC, KU Leuven, Leuven, Belgium.
| |
Collapse
|