1
|
Ximenes-Carballo C, Rey-Viñolas S, Blanco-Fernandez B, Pérez-Amodio S, Engel E, Castano O. Combining three-dimensionality and CaP glass-PLA composites: Towards an efficient vascularization in bone tissue healing. BIOMATERIALS ADVANCES 2024; 164:213985. [PMID: 39146606 DOI: 10.1016/j.bioadv.2024.213985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Bone regeneration often fails due to implants/grafts lacking vascular supply, causing necrotic tissue and poor integration. Microsurgical techniques are used to overcome this issue, allowing the graft to anastomose. These techniques have limitations, including severe patient morbidity and current research focuses on stimulating angiogenesis in situ using growth factors, presenting limitations, such as a lack of control and increased costs. Non-biological stimuli are necessary to promote angiogenesis for successful bone constructs. Recent studies have reported that bioactive glass dissolution products, such as calcium-releasing nanoparticles, stimulate hMSCs to promote angiogenesis and new vasculature. Moreover, the effect of 3D microporosity has also been reported to be important for vascularisation in vivo. Therefore, we used room-temperature extrusion 3D printing with polylactic acid (PLA) and calcium phosphate (CaP) based glass scaffolds, focusing on geometry and solvent displacement for scaffold recovery. Combining both methods enabled reproducible control of 3D structure, porosity, and surface topography. Scaffolds maintained calcium ion release at physiological levels and supported human mesenchymal stem cell proliferation. Scaffolds stimulated the secretion of vascular endothelial growth factor (VEGF) after 3 days of culture. Subcutaneous implantation in vivo indicated good scaffold integration and blood vessel infiltration as early as one week after. PLA-CaP scaffolds showed increased vessel maturation 4 weeks after implantation without vascular regression. Results show PLA/CaP-based glass scaffolds, made via controlled 3D printing, support angiogenesis and vessel maturation, promising improved vascularization for bone regeneration.
Collapse
Affiliation(s)
- Celia Ximenes-Carballo
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Sergi Rey-Viñolas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Barbara Blanco-Fernandez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Soledad Pérez-Amodio
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; IMEM-BRT group, Materials Science and Engineering, Polytechnical University of Catalonia (UPC), Barcelona, Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; IMEM-BRT group, Materials Science and Engineering, Polytechnical University of Catalonia (UPC), Barcelona, Spain.
| | - Oscar Castano
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain; Nanobioengineering and Biomaterials, Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.
| |
Collapse
|
2
|
Çelik M, Karaduman ZO, Turhan Y, Arıcan M, Gamsızkan M, Saglam S, Uludag V. The Effects of Phenyramidol and Diclofenac Treatment on Fracture Healing in Rats. Clin Orthop Surg 2024; 16:836-844. [PMID: 39364103 PMCID: PMC11444952 DOI: 10.4055/cios24056] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 10/05/2024] Open
Abstract
Background Fracture healing or nonunion refers to a process in which many factors interact. In this study, we aimed to evaluate the radiological, histological, and biomechanical effects of phenyramidol and diclofenac, which are frequently used to treat post-fracture ture pain worldwide, on fracture healing and nonunion in a rat femur fracture model. Methods In this study, 72 male Wistar-Albino rats aged 2-3 months and weighing 250 ± 30 g were divided into 4 main groups. The rats were divided into 12 subgroups according to the early, middle, and late periods. A fracture model was created in rat femurs, and surgical fixation was performed. Postoperative analgesic treatment protocols included phenyramidol, diclofenac, phenyramidol + diclofenac, and the control group. The rats were sacrificed on the fifteenth, thirtieth, and forty-fifth days and were evaluated radiologically, histopathologically, and biomechanically. Results Scoring was conducted independently by 2 orthopedists not involved in the study. When the results were analyzed statistically, no statistically significant difference was observed between the fifteenth and thirtieth day radiology score values of the control, diclofenac, phenyramidol, and Phenyramidol + diclofenac groups (p > 0.05), but there was a statistically significant difference (p < 0.05) between the forty-fifth day radiology score values of the control, diclofenac, phenyramidol, and phenyramidol + diclofenac groups. Conclusions Our study shows that the use of diclofenac or phenyramidol alone negatively affects postoperative fracture healing. However, this effect was less pronounced in the combined treatment group. Histologic examination revealed that neither treatment had a significant effect on healing. There were statistical differences in biomechanical and radiologic properties between the phenyramidol and diclofenac groups; in particular, the diclofenac group had lower biomechanical properties.
Collapse
Affiliation(s)
- Mücahit Çelik
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Düzce, Türkiye
| | - Zekeriya Okan Karaduman
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Düzce, Türkiye
| | - Yalcın Turhan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Düzce, Türkiye
| | - Mehmet Arıcan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Düzce, Türkiye
| | - Mehmet Gamsızkan
- Department of Pathology, Faculty of Medicine, Duzce University, Düzce, Türkiye
| | - Sönmez Saglam
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Düzce, Türkiye
| | - Veysel Uludag
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Duzce University, Düzce, Türkiye
| |
Collapse
|
3
|
Zhou H, Liu H, Lin M, Wang H, Zhou J, Li M, Yang X, Fu G, Liu C. Hyperbaric oxygen promotes bone regeneration by activating the mechanosensitive Piezo1 pathway in osteogenic progenitors. J Orthop Translat 2024; 48:11-24. [PMID: 39170748 PMCID: PMC11338066 DOI: 10.1016/j.jot.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/17/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Background Hyperbaric oxygen (HBO) therapy is widely used to treat bone defects, but the correlation of high oxygen concentration and pressure to osteogenesis is unclear. Methods Bilateral monocortical tibial defect surgeries were performed on 12-week-old Prrx1-Cre; Rosa26-tdTomato and Prrx1-Cre; Piezo1fl/+ mice. Daily HBO treatment was applied on post-surgery day (PSD) 1-9; and daily mechanical loading on tibia was from PSD 5 to 8. The mice were euthanized on PSD 10, and bone defect repair in their tibias was evaluated using μCT, biomechanical testing, and immunofluorescence deep-tissue imaging. The degree of angiogenesis-osteogenesis coupling was determined through spatial correlation analysis. Bone marrow stromal cells from knockout mice were cultured in vitro, and their osteogenic capacities of the cells were assessed. The activation of genes in the Piezo1-YAP pathway was evaluated using RNA sequencing and quantitative real-time polymerase chain reaction. Results Lineage tracing showed HBO therapy considerably altered the number of Prrx1+ cells and their progeny in a healing bone defect. Using conditional knockdown mice, we found that HBO stimulation activates the Piezo1-YAP axis in Prrx1+ cells and promotes osteogenesis-angiogenesis coupling during bone repair. The beneficial effect of HBO was similar to that of anabolic mechanical stimulation, which also acts through the Piezo1-YAP axis. Subsequent transcriptome sequencing results revealed that similar mechanosensitive pathways are activated by HBO therapy in a bone defect. Conclusion HBO therapy promotes bone tissue regeneration through the mechanosensitive Piezo1-YAP pathway in a population of Prrx1+ osteogenic progenitors. Our results contribute to the understanding of the mechanism by which HBO therapy treats bone defects. The Translational Potential of this Article Hyperbaric oxygen therapy is widely used in clinical settings. Our results show that osteogenesis was induced by the activation of the Piezo1-YAP pathway in osteoprogenitors after HBO stimulation, and the underlying mechanism was elucidated. These results may help improve current HBO methods and lead to the formulation of alternative treatments that achieve the same functional outcomes.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Hongzhi Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Hantang Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Jingjing Zhou
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Ming Li
- Department of Rehabilitation Medicine, Shenzhen Children's Hospital, No. 7019 Yitian Road, Futian District, Shenzhen, Guangdong, China
| | - Xue Yang
- Department of Rehabilitation Medicine, Shenzhen Children's Hospital, No. 7019 Yitian Road, Futian District, Shenzhen, Guangdong, China
| | - Guibing Fu
- Department of Pediatric Orthopedics, Shenzhen Children's Hospital, No. 7019 Yitian Road, Futian District, Shenzhen, Guangdong, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Morawska-Kozłowska M, Wilkosz A, Zhalniarovich Y. The Omentum-A Forgotten Structure in Veterinary Surgery in Small Animals' Surgery. Animals (Basel) 2024; 14:1848. [PMID: 38997960 PMCID: PMC11240631 DOI: 10.3390/ani14131848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
The greater and lesser omentum are derived from embryonic mesogastrium. The expansive greater omentum in dogs covers intestinal coils, while in cats, it is smaller. Comprising distinct portions, the greater omentum is rich in lymphatics and blood vessels. Conversely, the lesser omentum spans the liver, stomach, and duodenum. Studies on canine omentum reveal unique immune cell composition and regenerative potential attributed to adipose tissue-derived stromal cells (ADSCs). These cells hold promise in regenerative medicine, showing enhanced abilities compared with ADSCs from other sources. The omentum is critical in tissue repair and pathology, making it invaluable in veterinary surgery across various medical fields. The aim of this article was to research current knowledge about the applications of the omentum in veterinary surgery and the possibilities of using this structure in the future.
Collapse
Affiliation(s)
- Magdalena Morawska-Kozłowska
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Aleksandra Wilkosz
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Yauheni Zhalniarovich
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| |
Collapse
|
5
|
Gunes D, Oksuz S, Koseoglu RD, Gokce E. Comparison of the Effect of Platelet-rich Plasma (PRP) and Fat Graft on Autologous Bone Grafting in a Randomized-controlled Experimental Skull Model. J Craniofac Surg 2024; 35:1298-1304. [PMID: 38710066 DOI: 10.1097/scs.0000000000010166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/20/2024] [Indexed: 05/08/2024] Open
Abstract
Gold standard method for the treatment of critical-sized bone defects is the autogenous bone grafting procedure. A number of new and potentially useful adjuncts currently are being investigated to enhance the success of bone grafting. We propose to evaluate the effect of the most known and easily obtained 2 biological materials, fat graft and platelet-rich plasma (PRP), on bone graft healing. Twenty-seven New Zealand male rabbits were included in this randomized, controlled study. Two-sided 15-mm diameter bone defects were created in the parietal bones and the bones taken were replaced right-to-left and vice versa with 1 control group, 1 fat graft applied group, and the last one PRP applied group. Histologic evaluation and 3-dimensional maxillofacial computerized tomography were performed and bone density was calculated. In radiologic analysis, bone density was significantly different in the PRP group compared with the control and fat graft group in the 12th week ( P <0.05). In histologic scoring analysis, the PRP group had a better score than the control and fat graft group, while the fat graft group was worse than the control group in the 6th week ( P <0.05). The addition of PRP had a positive effect whereas fat graft had a negative effect on bone graft healing compared with the control group.
Collapse
Affiliation(s)
- Deniz Gunes
- Plastic, Reconstructive and Aesthetic Surgery Clinic, Aydin State Hospital, Aydin
| | - Sinan Oksuz
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Health Sciences, Gulhane Medical School, Ankara
| | | | - Erkan Gokce
- Department of Radiology, Gaziosmanpasa University, School of Medicine, Tokat, Turkey
| |
Collapse
|
6
|
Hughes L, Centner C. Idiosyncratic bone responses to blood flow restriction exercise: new insights and future directions. J Appl Physiol (1985) 2024; 136:283-297. [PMID: 37994414 PMCID: PMC11212818 DOI: 10.1152/japplphysiol.00723.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023] Open
Abstract
Applying blood flow restriction (BFR) during low-load exercise induces beneficial adaptations of the myotendinous and neuromuscular systems. Despite the low mechanical tension, BFR exercise facilitates a localized hypoxic environment and increase in metabolic stress, widely regarded as the primary stimulus for tissue adaptations. First evidence indicates that low-load BFR exercise is effective in promoting an osteogenic response in bone, although this has previously been postulated to adapt primarily during high-impact weight-bearing exercise. Besides studies investigating the acute response of bone biomarkers following BFR exercise, first long-term trials demonstrate beneficial adaptations in bone in both healthy and clinical populations. Despite the increasing number of studies, the physiological mechanisms are largely unknown. Moreover, heterogeneity in methodological approaches such as biomarkers of bone metabolism measured, participant and study characteristics, and time course of measurement renders it difficult to formulate accurate conclusions. Furthermore, incongruity in the methods of BFR application (e.g., cuff pressure) limits the comparability of datasets and thus hinders generalizability of study findings. Appropriate use of biomarkers, effective BFR application, and befitting study design have the potential to progress knowledge on the acute and chronic response of bone to BFR exercise and contribute toward the development of a novel strategy to protect or enhance bone health. Therefore, the purpose of the present synthesis review is to 1) evaluate current mechanistic evidence; 2) discuss and offer explanations for similar and contrasting data findings; and 3) create a methodological framework for future mechanistic and applied research.
Collapse
Affiliation(s)
- Luke Hughes
- Department of Sport Exercise & Rehabilitation, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Christoph Centner
- Department of Sport and Sport Science, University of Freiburg, Freiburg, Germany
- Praxisklinik Rennbahn, Muttenz, Switzerland
| |
Collapse
|
7
|
Menger MM, Bleimehl M, Bauer D, Scheuer C, Hans S, Saul D, Ehnert S, Menger MD, Histing T, Laschke MW. Cilostazol promotes blood vessel formation and bone regeneration in a murine non-union model. Biomed Pharmacother 2023; 168:115697. [PMID: 37864892 DOI: 10.1016/j.biopha.2023.115697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/23/2023] Open
Abstract
Non-unions represent a major complication in trauma and orthopedic surgery. Many factors contribute to bone regeneration, out of which an adequate vascularization has been recognized as crucial. The phosphodiesterase-3 (PDE-3) inhibitor cilostazol has been shown to exert pro-angiogenic and pro-osteogenic effects in a variety of preclinical studies. Hence, we herein investigated the effects of cilostazol on bone regeneration in an atrophic non-union model in mice. For this purpose, a 1.8 mm femoral segmental defect was stabilized by pin-clip fixation and the animals were treated daily with 30 mg/kg body weight cilostazol or saline (control) per os. At 2, 5 and 10 weeks after surgery the healing of femora was analyzed by X-ray, biomechanics, photoacoustic imaging, and micro-computed tomography (µCT). To investigate the cellular composition and the growth factor expression of the callus tissue additional histological, immunohistochemical and Western blot analyses were performed. Cilostazol-treated animals showed increased bone formation within the callus, resulting in an enhanced bending stiffness when compared to controls. This was associated with a more pronounced expression of vascular endothelial growth factor (VEGF), a higher number of CD31-positive microvessels and an increased oxygen saturation within the callus tissue. Furthermore, cilostazol induced higher numbers of tartrate-resistant acidic phosphate (TRAP)-positive osteoclasts and CD68-positive macrophages. Taken together, these findings demonstrate that cilostazol is a promising drug candidate for the adjuvant treatment of atrophic non-unions in clinical practice.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany; Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany.
| | - Michelle Bleimehl
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - David Bauer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Sandra Hans
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Dominik Saul
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Saar, Germany
| |
Collapse
|
8
|
Dazzi C, Mehl J, Benamar M, Gerhardt H, Knaus P, Duda GN, Checa S. External mechanical loading overrules cell-cell mechanical communication in sprouting angiogenesis during early bone regeneration. PLoS Comput Biol 2023; 19:e1011647. [PMID: 37956208 PMCID: PMC10681321 DOI: 10.1371/journal.pcbi.1011647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/27/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Sprouting angiogenesis plays a key role during bone regeneration. For example, insufficient early revascularization of the injured site can lead to delayed or non-healing. During sprouting, endothelial cells are known to be mechano-sensitive and respond to local mechanical stimuli. Endothelial cells interact and communicate mechanically with their surroundings, such as outer-vascular stromal cells, through cell-induced traction forces. In addition, external physiological loads act at the healing site, resulting in tissue deformations and impacting cellular arrangements. How these two distinct mechanical cues (cell-induced and external) impact angiogenesis and sprout patterning in early bone healing remains however largely unknown. Therefore, the aim of this study was to investigate the relative role of externally applied and cell-induced mechanical signals in driving sprout patterning at the onset of bone healing. To investigate cellular self-organisation in early bone healing, an in silico model accounting for the mechano-regulation of sprouting angiogenesis and stromal cell organization was developed. Computer model predictions were compared to in vivo experiments of a mouse osteotomy model stabilized with a rigid or a semirigid fixation system. We found that the magnitude and orientation of principal strains within the healing region can explain experimentally observed sprout patterning, under both fixation conditions. Furthermore, upon simulating the selective inhibition of either cell-induced or externally applied mechanical cues, external mechanical signals appear to overrule the mechanical communication acting on a cell-cell interaction level. Such findings illustrate the relevance of external mechanical signals over the local cell-mediated mechanical cues and could be used in the design of fracture treatment strategies for bone regeneration.
Collapse
Affiliation(s)
- Chiara Dazzi
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Mehl
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Mounir Benamar
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
9
|
Jin L, Long Y, Zhang Q, Long J. MiRNAs regulate cell communication in osteogenesis-angiogenesis coupling during bone regeneration. Mol Biol Rep 2023; 50:8715-8728. [PMID: 37642761 DOI: 10.1007/s11033-023-08709-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023]
Abstract
Bone regeneration is a complex process that requires not only the participation of multiple cell types, but also signal communication between cells. The two basic processes of osteogenesis and angiogenesis are closely related to bone regeneration and bone homeostasis. H-type vessels are a subtype of bone vessels characterized by high expression of CD31 and EMCN. These vessels play a key role in the regulation of bone regeneration and are important mediators of coupling between osteogenesis and angiogenesis. Molecular regulation between different cell types is important for coordination of osteogenesis and angiogenesis that promotes bone regeneration. MiRNAs are small non-coding RNAs that predominantly regulate gene expression at the post-transcriptional level and are closely related to cell communication. Specifically, miRNAs transduce external stimuli through various cell signaling pathways and cause a series of physiological and pathological effects. They are also deeply involved in the bone repair process. This review focuses on three signaling pathways related to osteogenesis-angiogenesis coupling, as well as the miRNAs involved in these pathways. Elucidation of the molecular mechanisms governing osteogenesis and angiogenesis is of great significance for bone regeneration.
Collapse
Affiliation(s)
- Liangyu Jin
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Yifei Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Qiuling Zhang
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Jie Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China.
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China.
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
10
|
Menger MM, Bauer D, Bleimehl M, Scheuer C, Braun BJ, Herath SC, Rollmann MF, Menger MD, Laschke MW, Histing T. Sildenafil, a phosphodiesterase-5 inhibitor, stimulates angiogenesis and bone regeneration in an atrophic non-union model in mice. J Transl Med 2023; 21:607. [PMID: 37684656 PMCID: PMC10486066 DOI: 10.1186/s12967-023-04441-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/14/2023] [Indexed: 09/10/2023] Open
Abstract
Non-union formation represents a major complication in trauma and orthopedic surgery. The phosphodiesterase-5 (PDE-5) inhibitor sildenafil has been shown to exert pro-angiogenic and pro-osteogenic effects in vitro and in vivo. Therefore, the aim of the present study was to analyze the impact of sildenafil in an atrophic non-union model in mice. After creation of a 1.8 mm segmental defect, mice femora were stabilized by pin-clip fixation. Bone regeneration was analyzed by means of X-ray, biomechanics, photoacoustic and micro-computed tomography (µCT) imaging as well as histological, immunohistochemical and Western blot analyses at 2, 5 and 10 weeks after surgery. The animals were treated daily with either 5 mg/kg body weight sildenafil (n = 35) or saline (control; n = 35) per os. Bone formation was markedly improved in defects of sildenafil-treated mice when compared to controls. This was associated with a higher bending stiffness as well as an increased number of CD31-positive microvessels and a higher oxygen saturation within the callus tissue. Moreover, the bone defects of sildenafil-treated animals contained more tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts and CD68-positive macrophages and exhibited a higher expression of the pro-angiogenic and pro-osteogenic markers cysteine rich protein (CYR)61 and vascular endothelial growth factor (VEGF) when compared to controls. These findings demonstrate that sildenafil acts as a potent stimulator of angiogenesis and bone regeneration in atrophic non-unions.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany.
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| | - David Bauer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Michelle Bleimehl
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Claudia Scheuer
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Benedikt J Braun
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Steven C Herath
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Mika F Rollmann
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076, Tuebingen, Germany
| |
Collapse
|
11
|
Roldan L, Isaza C, Ospina J, Montoya C, Domínguez J, Orrego S, Correa S. A Comparative Study of HA/DBM Compounds Derived from Bovine and Porcine for Bone Regeneration. J Funct Biomater 2023; 14:439. [PMID: 37754853 PMCID: PMC10532284 DOI: 10.3390/jfb14090439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/25/2023] [Accepted: 08/17/2023] [Indexed: 09/28/2023] Open
Abstract
This comparative study investigated the tissue regeneration and inflammatory response induced by xenografts comprised of hydroxyapatite (HA) and demineralized bone matrix (DBM) extracted from porcine (P) and bovine (B) sources. First, extraction of HA and DBM was independently conducted, followed by chemical and morphological characterization. Second, mixtures of HA/DBM were prepared in 50/50 and 60/40 concentrations, and the chemical, morphological, and mechanical properties were evaluated. A rat calvarial defect model was used to evaluate the tissue regeneration and inflammatory responses at 3 and 6 months. The commercial allograft DBM Puros® was used as a clinical reference. Different variables related to tissue regeneration were evaluated, including tissue thickness regeneration (%), amount of regenerated bone area (%), and amount of regenerated collagen area (%). The inflammatory response was evaluated by quantifying the blood vessel area. Overall, tissue regeneration from porcine grafts was superior to bovine. After 3 months of implantation, the tissue thickness regeneration in the 50/50P compound and the commercial DBM was significantly higher (~99%) than in the bovine materials (~23%). The 50/50P and DBM produced higher tissue regeneration than the naturally healed controls. Similar trends were observed for the regenerated bone and collagen areas. The blood vessel area was correlated with tissue regeneration in the first 3 months of evaluation. After 6 months of implantation, HA/DBM compounds showed less regenerated collagen than the DBM-only xenografts. In addition, all animal-derived xenografts improved tissue regeneration compared with the naturally healed defects. No clinical complications associated with any implanted compound were noted.
Collapse
Affiliation(s)
- Lina Roldan
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
| | - Catalina Isaza
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
| | - Juan Ospina
- Centro de Investigación y Desarrollo Cárnico, Industrias de Alimentos Zenú S.A.S., Grupo Nutresa, Medellín 050044, Colombia;
| | - Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
| | - José Domínguez
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA 191122, USA
| | - Santiago Correa
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
- Escuela de Ciencias Aplicadas e Ingeniería, Universidad EAFIT, Medellín 050022, Colombia
| |
Collapse
|
12
|
Alharbi MA, Graves DT. FOXO 1 deletion in chondrocytes rescues diabetes-impaired fracture healing by restoring angiogenesis and reducing apoptosis. Front Endocrinol (Lausanne) 2023; 14:1136117. [PMID: 37576976 PMCID: PMC10421747 DOI: 10.3389/fendo.2023.1136117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 06/12/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Diabetes mellitus is associated with higher risks of long bone and jaw fractures. It is also associated with a higher incidence of delayed union or non-union. Our previous investigations concluded that a dominant mechanism was the premature loss of cartilage during endochondral bone formation associated with increased osteoclastic activities. We tested the hypothesis that FOXO1 plays a key role in diabetes-impaired angiogenesis and chondrocyte apoptosis. Methods Closed fractures of the femur were induced in mice with lineage-specific FOXO1 deletion in chondrocytes. The control group consisted of mice with the FOXO1 gene present. Mice in the diabetic group were rendered diabetic by multiple streptozotocin injections, while mice in the normoglycemic group received vehicle. Specimens were collected 16 days post fracture. The samples were fixed, decalcified, and embedded in paraffin blocks for immunostaining utilizing anti cleaved caspase-3 or CD31 specific antibodies compared with matched control IgG antibody, and apoptosis by the TUNEL assay. Additionally, ATDC5 chondrocytes were examined in vitro by RT-PCR, luciferase reporter and chromatin immunoprecipitation assays. Results Diabetic mice had ~ 50% fewer blood vessels compared to normoglycemic mice FOXO1 deletion in diabetic mice partially rescued the low number of blood vessels (p < 0.05). Additionally, diabetes increased caspase-3 positive and apoptotic chondrocytes by 50%. FOXO1 deletion in diabetic animals blocked the increase in both to levels comparable to normoglycemic animals (p < 0.05). High glucose (HG) and high advanced glycation end products (AGE) levels stimulated FOXO1 association with the caspase-3 promoter in vitro, and overexpression of FOXO1 increased caspase-3 promoter activity in luciferase reporter assays. Furthermore, we review previous mechanistic studies demonstrating that tumor necrosis factor (TNF) inhibition reverses impaired angiogenesis and reverses high levels of chondrocyte apoptosis that occur in fracture healing. Discussion New results presented here, in combination with recent studies, provide a comprehensive overview of how diabetes, through high glucose levels, AGEs, and increased inflammation, impair the healing process by interfering with angiogenesis and stimulating chondrocyte apoptosis. FOXO1 in diabetic fractures plays a negative role by reducing new blood vessel formation and increasing chondrocyte cell death which is distinct from its role in normal fracture healing.
Collapse
Affiliation(s)
- Mohammed A. Alharbi
- Department of Endodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Dana T. Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
13
|
Watanabe H, Maishi N, Hoshi-Numahata M, Nishiura M, Nakanishi-Kimura A, Hida K, Iimura T. Skeletal-Vascular Interactions in Bone Development, Homeostasis, and Pathological Destruction. Int J Mol Sci 2023; 24:10912. [PMID: 37446097 DOI: 10.3390/ijms241310912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Bone is a highly vascularized organ that not only plays multiple roles in supporting the body and organs but also endows the microstructure, enabling distinct cell lineages to reciprocally interact. Recent studies have uncovered relevant roles of the bone vasculature in bone patterning, morphogenesis, homeostasis, and pathological bone destruction, including osteoporosis and tumor metastasis. This review provides an overview of current topics in the interactive molecular events between endothelial cells and bone cells during bone ontogeny and discusses the future direction of this research area to find novel ways to treat bone diseases.
Collapse
Affiliation(s)
- Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Nako Maishi
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Marie Hoshi-Numahata
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Mai Nishiura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Atsuko Nakanishi-Kimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| |
Collapse
|
14
|
Torrecillas-Baena B, Pulido-Escribano V, Dorado G, Gálvez-Moreno MÁ, Camacho-Cardenosa M, Casado-Díaz A. Clinical Potential of Mesenchymal Stem Cell-Derived Exosomes in Bone Regeneration. J Clin Med 2023; 12:4385. [PMID: 37445420 DOI: 10.3390/jcm12134385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Bone metabolism is regulated by osteoblasts, osteoclasts, osteocytes, and stem cells. Pathologies such as osteoporosis, osteoarthritis, osteonecrosis, and traumatic fractures require effective treatments that favor bone formation and regeneration. Among these, cell therapy based on mesenchymal stem cells (MSC) has been proposed. MSC are osteoprogenitors, but their regenerative activity depends in part on their paracrine properties. These are mainly mediated by extracellular vesicle (EV) secretion. EV modulates regenerative processes such as inflammation, angiogenesis, cell proliferation, migration, and differentiation. Thus, MSC-EV are currently an important tool for the development of cell-free therapies in regenerative medicine. This review describes the current knowledge of the effects of MSC-EV in the different phases of bone regeneration. MSC-EV has been used by intravenous injection, directly or in combination with different types of biomaterials, in preclinical models of bone diseases. They have shown great clinical potential in regenerative medicine applied to bone. These findings should be confirmed through standardization of protocols, a better understanding of the mechanisms of action, and appropriate clinical trials. All that will allow the translation of such cell-free therapy to human clinic applications.
Collapse
Affiliation(s)
- Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Victoria Pulido-Escribano
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Gabriel Dorado
- Department Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, 14071 Córdoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Marta Camacho-Cardenosa
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| |
Collapse
|
15
|
Irfan D, Ahmad I, Patra I, Margiana R, Rasulova MT, Sivaraman R, Kandeel M, Mohammad HJ, Al-Qaim ZH, Jawad MA, Mustafa YF, Ansari MJ. Stem cell-derived exosomes in bone healing: focusing on their role in angiogenesis. Cytotherapy 2023; 25:353-361. [PMID: 36241491 DOI: 10.1016/j.jcyt.2022.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/12/2022]
Abstract
Fractures in bone, a tissue critical in protecting other organs, affect patients' quality of life and have a heavy economic burden on societies. Based on regenerative medicine and bone tissue engineering approaches, stem cells have become a promising and attractive strategy for repairing bone fractures via differentiation into bone-forming cells and production of favorable mediators. Recent evidence suggests that stem cell-derived exosomes could mediate the therapeutic effects of their counterpart cells and provide a cell-free therapeutic strategy in bone repair. Since bone is a highly vascularized tissue, coupling angiogenesis and osteogenesis is critical in bone fracture healing; thus, developing therapeutic strategies to promote angiogenesis will facilitate bone regeneration and healing. To this end, stem cell-derived exosomes with angiogenic potency have been developed to improve fracture healing. This review summarizes the effects of stem cell-derived exosomes on the repair of bone tissue, focusing on the angiogenesis process.
Collapse
Affiliation(s)
- Daniyal Irfan
- School of Management, Guangzhou University, Guangzhou, China
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Depok, Indonesia; Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Depok, Indonesia; Dr Soetomo General Academic Hospital, Surabaya, Indonesia.
| | | | - R Sivaraman
- Department of Mathematics, Dwaraka Doss Goverdhan Doss Vaishnav College, University of Madras, Chennai, India
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelshikh University, Kafrelshikh, Egypt.
| | | | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
16
|
Dibazar ZE, Nie L, Azizi M, Nekounam H, Hamidi M, Shavandi A, Izadi Z, Delattre C. Bioceramics/Electrospun Polymeric Nanofibrous and Carbon Nanofibrous Scaffolds for Bone Tissue Engineering Applications. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2799. [PMID: 37049093 PMCID: PMC10095723 DOI: 10.3390/ma16072799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Bone tissue engineering integrates biomaterials, cells, and bioactive agents to propose sophisticated treatment options over conventional choices. Scaffolds have central roles in this scenario, and precisely designed and fabricated structures with the highest similarity to bone tissue have shown promising outcomes. On the other hand, using nanotechnology and nanomaterials as the enabling options confers fascinating properties to the scaffolds, such as precisely tailoring the physicochemical features and better interactions with cells and surrounding tissues. Among different nanomaterials, polymeric nanofibers and carbon nanofibers have attracted significant attention due to their similarity to bone extracellular matrix (ECM) and high surface-to-volume ratio. Moreover, bone ECM is a biocomposite of collagen fibers and hydroxyapatite crystals; accordingly, researchers have tried to mimic this biocomposite using the mineralization of various polymeric and carbon nanofibers and have shown that the mineralized nanofibers are promising structures to augment the bone healing process in the tissue engineering scenario. In this paper, we reviewed the bone structure, bone defects/fracture healing process, and various structures/cells/growth factors applicable to bone tissue engineering applications. Then, we highlighted the mineralized polymeric and carbon nanofibers and their fabrication methods.
Collapse
Affiliation(s)
- Zahra Ebrahimvand Dibazar
- Department of Oral and Maxillo Facial Medicine, Faculty of Dentistry, Tabriz Azad University of Medical Sciences, Tabriz 5165687386, Iran
| | - Lei Nie
- College of Life Sciences, Xinyang Normal University, Xinyang 464000, China
| | - Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan 6517838636, Iran
| | - Houra Nekounam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Masoud Hamidi
- Université Libre de Bruxelles (ULB), École Polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50-CP 165/61, 1050 Brussels, Belgium
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École Polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50-CP 165/61, 1050 Brussels, Belgium
| | - Zhila Izadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6714869914, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah 6714869914, Iran
| | - Cédric Delattre
- Clermont Auvergne INP, CNRS, Institut Pascal, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
- Institut Universitaire de France (IUF), 1 Rue Descartes, 75005 Paris, France
| |
Collapse
|
17
|
Buettmann EG, DeNapoli RC, Abraham LB, Denisco JA, Lorenz MR, Friedman MA, Donahue HJ. Reambulation following hindlimb unloading attenuates disuse-induced changes in murine fracture healing. Bone 2023; 172:116748. [PMID: 37001629 DOI: 10.1016/j.bone.2023.116748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Patients with bone and muscle loss from prolonged disuse have higher risk of falls and subsequent fragility fractures. In addition, fracture patients with continued disuse and/or delayed physical rehabilitation have worse clinical outcomes compared to individuals with immediate weight-bearing activity following diaphyseal fracture. However, the effects of prior disuse followed by physical reambulation on fracture healing cellular processes and adjacent bone and skeletal muscle recovery post-injury remains poorly defined. To bridge this knowledge gap and inform future treatment and rehabilitation strategies for fractures, a preclinical model of fracture healing with a history of prior unloading with and without reambulation was employed. First, skeletally mature male and female C57BL/6J mice (18 weeks) underwent hindlimb unloading by tail suspension (HLU) for 3 weeks to induce significant bone and muscle loss modeling enhanced bone fragility. Next, mice had their right femur fractured by open surgical dissection (stabilized with 24-gauge pin). The, mice were randomly assigned to continued HLU or allowed normal weight-bearing reambulation (HLU + R). Mice given normal cage activity throughout the experiment served as healthy age-matched controls. All mice were sacrificed 4-days (DPF4) or 14-days (DPF14) following fracture to assess healing and uninjured hindlimb musculoskeletal properties (6-10 mice per treatment/biological sex). We found that continued disuse following fracture lead to severely diminished uninjured hindlimb skeletal muscle mass (gastrocnemius and soleus) and femoral bone volume adjacent to the fracture site compared to healthy age-matched controls across mouse sexes. Furthermore, HLU led to significantly decreased periosteal expansion (DPF4) and osteochondral tissue formation by DPF14, and trends in increased osteoclastogenesis (DPF14) and decreased woven bone vascular area (DPF14). In contrast, immediate reambulation for 2 weeks after fracture, even following a period of prolonged disuse, was able to increase hindlimb skeletal tissue mass and increase osteochondral tissue formation, albeit not to healthy control levels, in both mouse sexes. Furthermore, reambulation attenuated osteoclast formation seen in woven bone tissue undergoing disuse. Our results suggest that weight-bearing skeletal loading in both sexes immediately following fracture may improve callus healing and prevent further fall risk by stimulating skeletal muscle anabolism and decreasing callus resorption compared to minimal or delayed rehabilitation regimens.
Collapse
Affiliation(s)
- Evan G Buettmann
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Rachel C DeNapoli
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Lovell B Abraham
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Joe A Denisco
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Madelyn R Lorenz
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Michael A Friedman
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Henry J Donahue
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America.
| |
Collapse
|
18
|
Li Y, Liu L, Li Y, Song W, Shao B, Li H, Lin W, Li Q, Shuai X, Bai M, Zhao B, Guo Y, Yuan Q, Wang Y. Alpha-ketoglutarate promotes alveolar bone regeneration by modulating M2 macrophage polarization. Bone Rep 2023; 18:101671. [PMID: 37007218 PMCID: PMC10064115 DOI: 10.1016/j.bonr.2023.101671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Objectives Alpha-ketoglutarate (αKG) is an essential metabolite that plays a crucial role in skeletal homeostasis. Here we aim to investigate the effect of αKG on alveolar socket healing and reveal the underlying mechanism in the view of macrophage polarization. Methods In a murine model pretreated with or without αKG, mandibular first molars were extracted. Mandibular tissues were harvested for microCT and histological analyses. Immunofluorescence was used to evaluate macrophage polarization during healing process. Macrophages with αKG/vehicle supplementation in vitro were proceeded to quantitative real-time PCR and flow cytometry to further elucidate the mechanism. Results MicroCT and histological analyses showed accelerated healing and enhanced bone regeneration of extraction sockets in experimental group. αKG increased new bone volume in alveolar sockets and promoted the activity of both osteoblastogenesis and osteoclastogenesis. αKG administration reduced M1 pro-inflammatory macrophages in an early phase and promoted anti-inflammatory M2 macrophage polarization in a later phase. Consistently, the expressions of M2 marker genes were augmented in αKG group, while M1 marker genes were downregulated. Flow cytometry revealed the increased ratio of M2/M1 macrophages in cells treated with αKG. Conclusions αKG accelerates the healing process of extraction sockets via orchestrating macrophage activation, with promising therapeutic potential in oral clinics.
Collapse
|
19
|
Chen Q, Wang Z, Yang C, Li B, Ren X, Liu C, Xi L. High resolution intravital photoacoustic microscopy reveals VEGF-induced bone regeneration in mouse tibia. Bone 2023; 167:116631. [PMID: 36435450 DOI: 10.1016/j.bone.2022.116631] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022]
Abstract
Osteogenesis and angiogenesis are essential for bone homeostasis and repair. Newly formed vessels convey osteogenic progenitors during bone regeneration. However, the lack of continuous and label-free visualization of the bone microvasculature has resulted in little understanding of the neovascular dynamics. Here, we take advantage of optical-resolution photoacoustic microscopy (ORPAM) for label-free, intravital, long-term observation of the bone vascular dynamics, including angiogenesis, remodeling and quantified angiogenic effect of locally-applied vascular endothelial growth factor (VEGF) in the murine tibial defect model. We employed ex vivo confocal microscopy and micro-computed tomography (micro-CT) imaging to verify the positive role of VEGF treatment. VEGF treatment increased the concentration of total hemoglobin, vascular branching, and vascular density, which correlated with more osteoprogenitors and increased bone formation within the defect. These data demonstrated ORPAM as a useful imaging tool that detected functional capillaries to understand hemodynamics, and revealed the effectiveness of locally delivered therapeutic agents with sufficient sensitivity, contributing to the understanding of spatiotemporal regulatory mechanisms on blood vessels during bone regeneration.
Collapse
Affiliation(s)
- Qian Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ziyan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Chengyu Yang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Baochen Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xingxing Ren
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| | - Lei Xi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Shenzhen Bay Laboratory, Shenzhen, Guangdong 518132, China.
| |
Collapse
|
20
|
Guo T, Yuan X, Li X, Liu Y, Zhou J. Bone regeneration of mouse critical-sized calvarial defects with human mesenchymal stem cell sheets co-expressing BMP2 and VEGF. J Dent Sci 2023; 18:135-144. [PMID: 36643246 PMCID: PMC9831827 DOI: 10.1016/j.jds.2022.06.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/20/2022] [Indexed: 01/18/2023] Open
Abstract
Background/purpose Over-dependence on existing synthetic scaffolds and insufficient osteoinductive and vasculogenic growth factors have limited the development of bone regeneration. The study aimed to assess the feasibility of using marrow-derived mesenchymal stem cells (BMSCs) cell sheets co-expressing bone morphogenetic proteins 2 (BMP2) and vascular endothelial growth factor (VEGF) for repairing critical-sized calvarial defects. Materials and methods BMSCs cell sheets were genetically engineered to express BMP2/VEGF alone or together. Alterations in osteogenic markers were examined by quantitative real-time PCR (qRT-PCR) and western blotting. A critical-sized calvarial bone defect model was used to investigate the osteogenesis effects of BMP2/VEGF cell sheets alone or in combination. The efficacy was assessed with micro-computed tomography (micro-CT) and histology. Results In vitro, the expression of BMP2 and VEGF through lentiviral transduction was confirmed by qRT-PCR and western blotting against BMP2 and VEGF. Lentiviral delivery of BMP2 and VEGF resulted in the upregulation of osteogenic markers. In vivo, in a critical-sized calvarial bone defect model, 3D-reconstructed micro-CT images revealed that treatment of the calvarial defects with the BMP2/VEGF cell sheet resulted in significantly greater amounts of newly formed bone at 8 weeks after surgery than treatment with cell sheets with single gene transduction or vehicle controls. The results were confirmed by histological assessment by H&E staining and Masson staining. Conclusion This study demonstrates that BMP2/VEGF co-expressing BMSCs sheets promote bone regeneration in critical-sized calvarial bone defects. The BMP2/VEGF cell sheets provide a functional bioactive scaffold for critical-size bone reconstruction.
Collapse
Affiliation(s)
- Tingting Guo
- Department of General Dentistry and Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing, PR China
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China
| | - Xiaohong Yuan
- Department of Pathology, Beijing Stomatological Hospital, Capital Medical University, Beijing, PR China
| | - Xin Li
- Department of Preventive Dentistry, Beijing Stomatological Hospital, Capital Medical University, Beijing, PR China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Jian Zhou
- Department of VIP Dental Service, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, PR China
| |
Collapse
|
21
|
Notermans T, Isaksson H. Predicting the formation of different tissue types during Achilles tendon healing using mechanoregulated and oxygen-regulated frameworks. Biomech Model Mechanobiol 2022; 22:655-667. [PMID: 36542228 PMCID: PMC10097799 DOI: 10.1007/s10237-022-01672-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
AbstractDuring Achilles tendon healing in rodents, besides the expected tendon tissue, also cartilage-, bone- and fat-like tissue features have been observed during the first twenty weeks of healing. Several studies have hypothesized that mechanical loading may play a key role in the formation of different tissue types during healing. We recently developed a computational mechanobiological framework to predict tendon tissue production, organization and mechanical properties during tendon healing. In the current study, we aimed to explore possible mechanobiological related mechanisms underlying formation of other tissue types than tendon tissue during tendon healing. To achieve this, we further developed our recent framework to predict formation of different tissue types, based on mechanobiological models established in other fields, which have earlier not been applied to study tendon healing. We explored a wide range of biophysical stimuli, i.e., principal strain, hydrostatic stress, pore pressure, octahedral shear strain, fluid flow, angiogenesis and oxygen concentration, that may promote the formation of different tissue types. The numerical framework predicted spatiotemporal formation of tendon-, cartilage-, bone- and to a lesser degree fat-like tissue throughout the first twenty weeks of healing, similar to recent experimental reports. Specific features of experimental data were captured by different biophysical stimuli. Our modeling approach showed that mechanobiology may play a role in governing the formation of different tissue types that have been experimentally observed during tendon healing. This study provides a numerical tool that can contribute to a better understanding of tendon mechanobiology during healing. Developing these tools can ultimately lead to development of better rehabilitation regimens that stimulate tendon healing and prevent unwanted formation of cartilage-, fat- and bone-like tissues.
Collapse
Affiliation(s)
- Thomas Notermans
- Department of Biomedical Engineering, Lund University, Lund, Sweden.
| | - Hanna Isaksson
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| |
Collapse
|
22
|
McKenzie JA, Galbreath IM, Coello AF, Hixon KR, Silva MJ. VEGFA from osteoblasts is not required for lamellar bone formation following tibial loading. Bone 2022; 163:116502. [PMID: 35872107 PMCID: PMC9624127 DOI: 10.1016/j.bone.2022.116502] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/05/2022] [Accepted: 07/18/2022] [Indexed: 11/27/2022]
Abstract
The relationship between osteogenesis and angiogenesis is complex. Normal bone development requires angiogenesis, mediated by vascular endothelial growth factor A (VEGFA). Studies have demonstrated through systemic inhibition or genetic modification that VEGFA is indispensable for several types of bone repair, presumably via its role in supporting angiogenesis. But a direct role for VEGFA within osteoblasts, in the absence of angiogenesis, has also been suggested. To address the question of whether VEGFA from osteoblasts supports bone formation directly, we applied anabolic loading to induce lamellar bone formation in mice, a process shown to be independent of angiogenesis. We hypothesized that VEGFA from osteoblasts is required for lamellar bone formation. To test this hypothesis, we applied axial tibial compression to inducible Cre/LoxP mice from three lines. Vegfafl/fl mice were crossed with Ubiquitin C (UBC), Osterix (Osx) and Dentin-Matrix Protein 1 (DMP1) Cre-ERT2 mice to target all cells, (pre)osteoblast-lineage cells, and mature osteoblasts and osteocytes, respectively. Genotype effects were determined by comparing control (Vegfafl/fl) and Cre+ (VegfaΔ) mice for each line. At 5 months of age tamoxifen was injected for 5 days followed by a 3-week clearance prior to loading. Female and male mice (N = 100) were loaded for 5 days to peak forces to engender -3100 με peak compressive strain and processed for dynamic histomorphometry (day 12). Percent MS/BS increased 20-70 % as a result of loading, with no effect of genotype in Osx or Dmp1 lines. In contrast, the UBC groups had a significant decrease in relative periosteal BFR/BS in VegfaΔ vs. Vegfafl/fl mice. The UBC line did not have any cortical bone phenotype in non-loaded femurs. In summary, dynamic histomorphometry data confirmed that tibial loading induces lamellar bone formation. Contrary to our hypothesis, there was no decrease in loading-induced bone formation in the Osx or Dmp1 lines in the absence of VEGFA. There was a decrease in bone formation in the UBC line where all cells were targeted. This result indicates that VEGFA from a non-osteoblast cell source supports loading-induced lamellar bone formation, although osteoblast/osteocyte VEGFA is dispensable. These findings support a paracrine model whereby non-osteoblast VEGFA supports lamellar bone formation, independent of angiogenesis.
Collapse
Affiliation(s)
- Jennifer A McKenzie
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, MO, United States of America
| | - Ian M Galbreath
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, MO, United States of America; St. Louis University, St. Louis, MO, United States of America
| | - Andre F Coello
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States of America
| | - Katherine R Hixon
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, MO, United States of America; Dartmouth Engineering, Dartmouth College, Hanover, NH, United States of America
| | - Matthew J Silva
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States of America.
| |
Collapse
|
23
|
Zhang S, Tuk B, van de Peppel J, Kremers GJ, Koedam M, Pesch GR, Rahman Z, Hoogenboezem RM, Bindels EMJ, van Neck JW, Boukany PE, van Leeuwen JPTM, van der Eerden BCJ. Microfluidic evidence of synergistic effects between mesenchymal stromal cell-derived biochemical factors and biomechanical forces to control endothelial cell function. Acta Biomater 2022; 151:346-359. [PMID: 35995408 DOI: 10.1016/j.actbio.2022.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022]
Abstract
A functional vascular system is a prerequisite for bone repair as disturbed angiogenesis often causes non-union. Paracrine factors released from human bone marrow derived mesenchymal stromal cells (BMSCs) have angiogenic effects on endothelial cells. However, whether these paracrine factors participate in blood flow dynamics within bone capillaries remains poorly understood. Here, we used two different microfluidic designs to investigate critical steps during angiogenesis and found pronounced effects of endothelial cell proliferation as well as chemotactic and mechanotactic migration induced by BMSC conditioned medium (CM). The application of BMSC-CM in dynamic cultures demonstrates that bioactive factors in combination with fluidic flow-induced biomechanical signals significantly enhanced endothelial cell migration. Transcriptional analyses of endothelial cells demonstrate the induction of a unique gene expression profile related to tricarboxylic acid cycle and energy metabolism by the combination of BMSC-CM factors and shear stress, which opens an interesting avenue to explore during fracture healing. Our results stress the importance of in vivo - like microenvironments simultaneously including biochemical, biomechanical and oxygen levels when investigating key events during vessel repair. STATEMENT OF SIGNIFICANCE: Our results demonstrate the importance of recapitulating in vivo - like microenvironments when investigating key events during vessel repair. Endothelial cells exhibit enhanced angiogenesis characteristics when simultaneous exposing them to hMSC-CM, mechanical forces and biochemical signals simultaneously. The improved angiogenesis may not only result from the direct effect of growth factors, but also by reprogramming of endothelial cell metabolism. Moreover, with this model we demonstrated a synergistic impact of mechanical forces and biochemical factors on endothelial cell behavior and the expression of genes involved in the TCA cycle and energy metabolism, which opens an interesting new avenue to stimulate angiogenesis during fracture healing.
Collapse
Affiliation(s)
- Shuang Zhang
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bastiaan Tuk
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Jeroen van de Peppel
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Gert-Jan Kremers
- Erasmus Optical Imaging Center, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Marijke Koedam
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Georg R Pesch
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Eric M J Bindels
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Johan W van Neck
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Johannes P T M van Leeuwen
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bram C J van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands.
| |
Collapse
|
24
|
Rashidi A, Theruvath AJ, Huang CH, Wu W, Mahmoud EE, Jesu Raj JG, Marycz K, Daldrup-Link HE. Vascular injury of immature epiphyses impair stem cell engraftment in cartilage defects. Sci Rep 2022; 12:11696. [PMID: 35810189 PMCID: PMC9271080 DOI: 10.1038/s41598-022-15721-6] [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: 12/11/2021] [Accepted: 06/28/2022] [Indexed: 11/09/2022] Open
Abstract
The purpose of our study was to investigate if vascular injury in immature epiphyses affects cartilage repair outcomes of matrix-associated stem cell implants (MASI). Porcine bone marrow mesenchymal stromal stem cells (BMSCs) suspended in a fibrin glue scaffold were implanted into 24 full-thickness cartilage defects (5 mm ø) of the bilateral distal femur of six Göttingen minipigs (n = 12 defects in 6 knee joints of 3 immature pigs; age 3.5-4 months; n = 12 defects in 6 knee joints of 3 mature control pigs; age, 21-28 months). All pigs underwent magnetic resonance imaging (MRI) at 2, 4, 12 (n = 24 defects), and 24 weeks (n = 12 defects). After the last imaging study, pigs were sacrificed, joints explanted and evaluated with VEGF, H&E, van Gieson, Mallory, and Safranin O stains. Results of mature and immature cartilage groups were compared using the Wilcoxon signed-rank test. Quantitative scores for subchondral edema at 2 weeks were correlated with quantitative scores for cartilage repair (MOCART score and ICRS score) at 12 weeks as well as Pineda scores at end of the study, using linear regression analysis. On serial MRIs, mature joints demonstrated progressive healing of cartilage defects while immature joints demonstrated incomplete healing and damage of the subchondral bone. The MOCART score at 12 weeks was significantly higher for mature joints (79.583 ± 7.216) compared to immature joints (30.416 ± 10.543, p = 0.002). Immature cartilage demonstrated abundant microvessels while mature cartilage did not contain microvessels. Accordingly, cartilage defects in immature joints showed a significantly higher number of disrupted microvessels, subchondral edema, and angiogenesis compared to mature cartilage. Quantitative scores for subchondral edema at 2 weeks were negatively correlated with MOCART scores (r = - 0.861) and ICRS scores (r = - 0.901) at 12 weeks and positively correlated with Pineda scores at the end of the study (r = 0.782). Injury of epiphyseal blood vessels in immature joints leads to subchondral bone defects and limits cartilage repair after MASI.
Collapse
Affiliation(s)
- Ali Rashidi
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ashok J Theruvath
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ching-Hsin Huang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Wei Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Elhussein E Mahmoud
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA.,Department of Surgery, Veterinary School, South Valley University, Qena, Egypt
| | - Joe Gerald Jesu Raj
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Krzysztof Marycz
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA.,International Institute of Translational Medicine (MIMT), Malin, Wisznia Mała, Poland
| | - Heike E Daldrup-Link
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA. .,Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Jiang J, Röper L, Alageel S, Dornseifer U, Schilling AF, Hadjipanayi E, Machens HG, Moog P. Hypoxia Preconditioned Serum (HPS) Promotes Osteoblast Proliferation, Migration and Matrix Deposition. Biomedicines 2022; 10:biomedicines10071631. [PMID: 35884936 PMCID: PMC9313157 DOI: 10.3390/biomedicines10071631] [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: 06/06/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/21/2022] Open
Abstract
Interest in discovering new methods of employing natural growth factor preparations to promote bone fracture healing is becoming increasingly popular in the field of regenerative medicine. In this study, we were able to demonstrate the osteogenic potential of hypoxia preconditioned serum (HPS) on human osteoblasts in vitro. Human osteoblasts were stimulated with two HPS concentrations (10% and 40%) and subsequently analyzed at time points of days 2 and 4. In comparison to controls, a time- and dose-dependent (up to 14.2× higher) proliferation of osteoblasts was observed after 4 days of HPS-40% stimulation with lower lactate dehydrogenase (LDH)-levels detected than controls, indicating the absence of cytotoxic/stress effects of HPS on human osteoblasts. With regards to cell migration, it was found to be significantly faster with HPS-10% application after 72 h in comparison to controls. Further osteogenic response to HPS treatment was evaluated by employing culture supernatant analysis, which exhibited significant upregulation of OPG (Osteoprotegerin) with higher dosage (HPS-10% vs. HPS-40%) and longer duration (2 d vs. 4 d) of HPS stimulation. There was no detection of anti-osteogenic sRANKL (soluble Receptor Activator of NF-κB Ligand) after 4 days of HPS stimulation. In addition, ALP (alkaline phosphatase)-enzyme activity, was found to be upregulated, dose-dependently, after 4 days of HPS-40% application. When assessing ossification through Alizarin-Red staining, HPS dose-dependently achieved greater (up to 2.8× higher) extracellular deposition of calcium-phosphate with HPS-40% in comparison to controls. These findings indicate that HPS holds the potential to accelerate bone regeneration by osteogenic promotion of human osteoblasts.
Collapse
Affiliation(s)
- Jun Jiang
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany; (J.J.); (L.R.); (S.A.); (E.H.)
| | - Lynn Röper
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany; (J.J.); (L.R.); (S.A.); (E.H.)
| | - Sarah Alageel
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany; (J.J.); (L.R.); (S.A.); (E.H.)
| | - Ulf Dornseifer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Isar Klinikum, D-80331 Munich, Germany;
| | - Arndt F. Schilling
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, Universitätsmedizin Göttingen, D-37075 Göttingen, Germany;
| | - Ektoras Hadjipanayi
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany; (J.J.); (L.R.); (S.A.); (E.H.)
| | - Hans-Günther Machens
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany; (J.J.); (L.R.); (S.A.); (E.H.)
- Correspondence: (H.-G.M.); (P.M.)
| | - Philipp Moog
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technische Universität München, D-81675 Munich, Germany; (J.J.); (L.R.); (S.A.); (E.H.)
- Correspondence: (H.-G.M.); (P.M.)
| |
Collapse
|
26
|
Shanmugavadivu A, Balagangadharan K, Selvamurugan N. Angiogenic and Osteogenic Effects of Flavonoids in Bone Regeneration. Biotechnol Bioeng 2022; 119:2313-2330. [PMID: 35718883 DOI: 10.1002/bit.28162] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/10/2022]
Abstract
Bone is a highly vascularised tissue that relies on a close spatial and temporal interaction between blood vessels and bone cells. As a result, angiogenesis is critical for bone formation and healing. The vascular system supports bone regeneration by delivering oxygen, nutrients, and growth factors, as well as facilitating efficient cell-cell contact. Most clinical applications of engineered bone grafts are hampered by insufficient vascularization after implantation. Over the last decade, a number of flavonoids have been reported to have osteogenic-angiogenic potential in bone regeneration because of their excellent bioactivity, low cost, availability, and minimal in vivo toxicity. During new bone formation, the osteoinductive nature of certain flavonoids is involved in regulating multiple signaling pathways contributing toward the osteogenic-angiogenic coupling. This review briefly outlines the osteogenic-angiogenic potential of those flavonoids and the mechanisms of their action in promoting bone regeneration. However, further studies are needed to investigate their delivery strategies and establish their clinical efficacy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - K Balagangadharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| |
Collapse
|
27
|
Fattahi R, Mohebichamkhorami F, Khani MM, Soleimani M, Hosseinzadeh S. Aspirin effect on bone remodeling and skeletal regeneration: Review article. Tissue Cell 2022; 76:101753. [PMID: 35180553 DOI: 10.1016/j.tice.2022.101753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/21/2022] [Accepted: 02/06/2022] [Indexed: 12/21/2022]
Abstract
Bone tissues are one of the most complex tissues in the body that regenerate and repair themselves spontaneously under the right physiological conditions. Within the limitations of treating bone defects, mimicking tissue engineering through the recruitment of scaffolds, cell sources and growth factors, is strongly recommended. Aspirin is one of the non-steroidal anti-inflammatory drugs (NSAIDs) and has been used in clinical studies for many years due to its anti-coagulant effect. On the other hand, aspirin and other NSAIDs activate cytokines and some mediators in osteoclasts, osteoblasts and their progenitor cells in a defect area, thereby promoting bone regeneration. It also stimulates angiogenesis by increasing migration of endothelial cells and the newly developed vessels are of emergency in bone fracture repair. This review covers the role of aspirin in bone tissue engineering and also, highlights its chemical reactions, mechanisms, dosages, anti-microbial and angiogenesis activities.
Collapse
Affiliation(s)
- Roya Fattahi
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Mohebichamkhorami
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Khani
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Simzar Hosseinzadeh
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Golestani S, Golestaneh A, Gohari AA. Comparative effects of systemic administration of levofloxacin and cephalexin on fracture healing in rats. J Korean Assoc Oral Maxillofac Surg 2022; 48:94-100. [PMID: 35491140 PMCID: PMC9065642 DOI: 10.5125/jkaoms.2022.48.2.94] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/02/2022] [Accepted: 03/15/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives This study aimed to compare the effects of systemic administration of levofloxacin or cephalexin on fracture healing in rats. Materials and Methods In this animal study, tibial fractures not requiring fixation were artificially induced in 30 male Wistar albino rats using a 1.1 mm surgical bur. The rats were randomly divided into 6 groups (n=5). Groups 1 and 2 received daily subcutaneous saline injections. Groups 3 and 4 received subcutaneous injections of 25 mg/kg levofloxacin twice daily. Groups 5 and 6 received daily subcutaneous injections of 20 mg/kg cephalexin. The rats in Groups 1, 3, and 5 were sacrificed after 1 week, while the rats in Groups 2, 4, and 6 were sacrificed after 4 weeks. The score of fracture healing was determined through histological assessment of sections from the fracture site according to Perry and colleagues. Data were analyzed by Kruskal-Wallis and Mann-Whitney tests. Results The mean score of fracture healing at 4 weeks was significantly higher than that at 1 week in the saline, levofloxacin, and cephalexin groups (P<0.001). At 1 week, no significant difference was noted among the three groups of saline, levofloxacin, and cephalexin in the mean score of fracture healing (P=0.360). However, this difference was significant at 4 weeks (P=0.018), and the mean score in the saline group was significantly higher compared to that in the levofloxacin group (P=0.015). Conclusion It is recommended not to prescribe levofloxacin for more than 1 week after surgical management of bone fractures due to its possible adverse effects on fracture healing.
Collapse
Affiliation(s)
- Shayan Golestani
- Department of Oral and Maxillofacial Surgery, Dental School, Islamic Azad University, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Arash Golestaneh
- Department of Oral and Maxillofacial Surgery, Dental School, Islamic Azad University, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Atousa Aminzadeh Gohari
- Department of Oral Pathology, Dental School, Islamic Azad University, Isfahan (Khorasgan) Branch, Isfahan, Iran
| |
Collapse
|
29
|
Wu S, Quan K, Wang W, Zhang Y, Mei J. 3D Mapping of Bone Channel of Blood Supply to Femoral Head in Proximal Femur. Front Surg 2022; 9:852653. [PMID: 36003280 PMCID: PMC9394460 DOI: 10.3389/fsurg.2022.852653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/12/2022] [Indexed: 11/28/2022] Open
Abstract
Background A detailed depiction of nutrient foramina is useful for defining guidelines and minimising iatrogenic damage during hip surgery. Therefore, this study aimed to define the location and frequency of nutrient foramina in the proximal femur using mapping techniques. Methods One hundred dry human cadaveric proximal femurs, comprising 56 left and 44 right femurs, were scanned using a three-dimensional scanner, with scanning distance 200 mm, precision 0.01 mm, and measuring point 0.04 mm. The image resolution of 1,310,000 pixels was obtained. Digital imaging models were acquired from the proximal femur surface. All the nutrient foramina in each model were identified and marked. The nutrient foramina models were superimposed on one another and oriented to fit a standard template of the femur’s proximal aspect. Three-dimensional mapping in the proximal femur’s nutrient foramina was performed. Results The nutrient foramina’s location and dense zones were identified. The dense zones were distributed along the vascular course and gaps between the muscle attachment sites. Eighteen dense zones were identified and found to be location-dependent. They were located in the central part of the fovea capitis femoris, subcapital and basicervical areas of the femoral neck, and muscle attachment gaps of the femoral trochanter. Conclusions The terminal branch of the nutrient vessels entering the nutrient foramina is at risk for iatrogenic damage during hip surgeries, especially in cases of close bone exposures. There are 18 dense zones that need to be considered for a safer approach to the proximal femur. To minimise iatrogenic damage to the nutrient vessels entering the nutrient foramina, the dense areas should be avoided when technically possible.
Collapse
Affiliation(s)
- Shenghui Wu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Kun Quan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Wei Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yingqi Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- Correspondence: Jiong Mei Yingqi Zhang
| | - Jiong Mei
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Correspondence: Jiong Mei Yingqi Zhang
| |
Collapse
|
30
|
Hixon KR, Miller AN. Animal models of impaired long bone healing and tissue engineering- and cell-based in vivo interventions. J Orthop Res 2022; 40:767-778. [PMID: 35072292 DOI: 10.1002/jor.25277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/05/2021] [Accepted: 01/16/2022] [Indexed: 02/04/2023]
Abstract
Bone healing after injury typically follows a systematic process and occurs spontaneously under appropriate physiological conditions. However, impaired long bone healing is still quite common and may require surgical intervention. Various complications can result in different forms of impaired bone healing including nonunion, critical-size defects, or stress fractures. While a nonunion may occur due to impaired biological signaling and/or mechanical instability, a critical-size defect exhibits extensive bone loss that will not spontaneously heal. Comparatively, a stress fracture occurs from repetitive forces and results in a non-healing crack or break in the bone. Clinical standards of treatment vary between these bone defects due to their pathological differences. The use of appropriate animal models for modeling healing defects is critical to improve current treatment methods and develop novel rescue therapies. This review provides an overview of these clinical bone healing impairments and current animal models available to study the defects in vivo. The techniques used to create these models are compared, along with the outcomes, to clarify limitations and future objectives. Finally, rescue techniques focused on tissue engineering and cell-based therapies currently applied in animal models are specifically discussed to analyze their ability to initiate healing at the defect site, providing information regarding potential future therapies. In summary, this review focuses on the current animal models of nonunion, critical-size defects, and stress fractures, as well as interventions that have been tested in vivo to provide an overview of the clinical potential and future directions for improving bone healing.
Collapse
Affiliation(s)
- Katherine R Hixon
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Thayer School of Engineering, Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Anna N Miller
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
31
|
Freischmidt H, Armbruster J, Rothhaas C, Titze N, Guehring T, Nurjadi D, Sonntag R, Schmidmaier G, Grützner PA, Helbig L. Treatment of Infection-Related Non-Unions with Bioactive Glass-A Promising Approach or Just Another Method of Dead Space Management? MATERIALS 2022; 15:ma15051697. [PMID: 35268930 PMCID: PMC8911496 DOI: 10.3390/ma15051697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 12/17/2022]
Abstract
The treatment of infected and non-infected non-unions remains a major challenge in trauma surgery. Due to the limited availability of autologous bone grafts and the need for local anti-infective treatment, bone substitutes have been the focus of tissue engineering for years. In this context, bioactive glasses are promising, especially regarding their anti-infective potential, which could reduce the need for local and systemic treatment with conventional antibiotics. The aim of this study was to investigate the osteoinductive and osteoconductive effects, as well as the anti-infectious potential, of S53P4 using a standardized non-union model, which had not been investigated previously. Using an already established sequential animal model in infected and non-infected rat femora, we were able to investigate bioactive glass S53P4 under realistic non-union conditions regarding its osteoinductive, osteoconductive and anti-infective potential with the use of µCT scans, biomechanical testing and histological, as well as microbiological, analysis. Although S53P4 did not lead to a stable union in the non-infected or the infected setting, µCT analysis revealed an osteoinductive effect of S53P4 under non-infected conditions, which was diminished under infected conditions. The osteoconductive effect of S53P4 remained almost negligible in histological analysis, even 8 weeks after treatment. Additionally, the expected anti-infective effect could not be demonstrated. Our data suggested that S53P4 should not be used in infected non-unions, especially in those with large bone defects.
Collapse
Affiliation(s)
- Holger Freischmidt
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Jonas Armbruster
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Catharina Rothhaas
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Nadine Titze
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Thorsten Guehring
- Trauma Centre, Hospital Paulinenhilfe Stuttgart at Tübingen University Hospital, Rosenbergstr. 38, 70176 Stuttgart, Germany;
| | - Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany;
| | - Robert Sonntag
- Laboratory of Biomechanics and Implant Research, Clinic for Orthopedics and Trauma Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany;
| | - Gerhard Schmidmaier
- Clinic for Orthopedics and Trauma Surgery, Center for Orthopedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany;
| | - Paul Alfred Grützner
- Department of Trauma and Orthopedic Surgery, BG Trauma Center Ludwigshafen at Heidelberg University Hospital, 67071 Ludwigshafen am Rhein, Germany; (H.F.); (J.A.); (C.R.); (N.T.); (P.A.G.)
| | - Lars Helbig
- Clinic for Orthopedics and Trauma Surgery, Center for Orthopedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany;
- Correspondence:
| |
Collapse
|
32
|
Zhou Z, Yan Y, Yu H, Yang G, Su H, Zhang T, Fan Y, Zhao F. Effect of Inter-Fragmentary Gap Size on Neovascularization During Bone Healing: A Micro-CT Imaging Study. Front Bioeng Biotechnol 2022; 10:808182. [PMID: 35345467 PMCID: PMC8957065 DOI: 10.3389/fbioe.2022.808182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/19/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction: Neovascularization of the fracture site is of great importance for bone healing and could be influenced by local mechanical environment such as fixation stability and inter-fragmentary gap size. This study aims to reconstruct the neovascularization of the fracture site and explore the effect of inter-fragmentary gap size on the spatiotemporal structure of vascularity during bone healing. Methods: Osteotomy was performed on 36 Sprague–Dawley (SD) rats on the right tibial diaphysis, and the fracture was given stable fixation with two different inter-fragmentary gap sizes. SD rats received stable fixation with either a small-sized inter-fragmentary gap (FSF1, 1 mm, n = 18) or a large-sized one (FSF3, 3 mm, n = 18). The left hind limbs were treated as the control group (CON). The animals were killed at different time points (2, 4, and 6 weeks postoperatively, n = 6, respectively) for vascular perfusion and micro-CT imaging. Results: (a) At week 2 and 4, FSF1 group showed significantly higher vessel volume ratio (VV/TV) and vessel surface density (VS/TV) values than both CON and FSF3 group; there was no significant difference in either VV/TV or VS/TV values between CON and FSF3 groups. (b) At week 6, both FSF1 and FSF3 groups showed significantly higher VV/TV and VS/TV values than CON group; FSF3 group had a significantly higher VV/TV value than FSF1 group. Conclusion: Different inter-fragmentary gap sizes greatly affect the timing of angiogenesis at the fracture site. Stable fixation with a small inter-fragmentary gap (1 mm) benefits neovascularization at the early stages during bone healing and reconstruction, while stable fixation with a large inter-fragmentary gap (3 mm) delays the occurrence of angiogenesis to a later phase.
Collapse
Affiliation(s)
- Zhilun Zhou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yang Yan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Hao Yu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Guanzhong Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Hao Su
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Tao Zhang
- Department of Orthopeadics, Tianjin Hospital, Tianjin, China
- *Correspondence: Tao Zhang, ; Yubo Fan, ; Feng Zhao,
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: Tao Zhang, ; Yubo Fan, ; Feng Zhao,
| | - Feng Zhao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: Tao Zhang, ; Yubo Fan, ; Feng Zhao,
| |
Collapse
|
33
|
Abstract
Despite major research efforts to elucidate mechanisms of non-union formation, failed fracture healing remains a common complication in orthopedic surgery. Adequate vascularization has been recognized as a crucial factor for successful bone regeneration, as newly formed microvessels guarantee the supply of the callus tissue with vital oxygen, nutrients, and growth factors. Accordingly, a vast number of preclinical studies have focused on the development of vascularization strategies to stimulate fracture repair. However, recent evidence suggests that stimulation of blood vessel formation is an oversimplified approach to support bone regeneration. This review discusses the role of vascularization during bone regeneration and delineates a phenomenon, for which we coin the term “the vascularization paradox of non-union-formation”. This view is based on the results of a variety of experimental studies that suggest that the callus tissue of non-unions is indeed densely vascularized and that pro-angiogenic mediators, such as vascular endothelial growth factor, are sufficiently expressed at the facture site. By gaining further insights into the molecular and cellular basis of non-union vascularization, it may be possible to develop more optimized treatment approaches or even prevent the non-union formation in the future.
Collapse
|
34
|
Bump RG, Goo CEA, Horton EC, Rasmussen JP. Osteoblasts pattern endothelium and somatosensory axons during zebrafish caudal fin organogenesis. Development 2022; 149:dev200172. [PMID: 35129199 PMCID: PMC8918783 DOI: 10.1242/dev.200172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022]
Abstract
Skeletal elements frequently associate with vasculature and somatosensory nerves, which regulate bone development and homeostasis. However, the deep, internal location of bones in many vertebrates has limited in vivo exploration of the neurovascular-bone relationship. Here, we use the zebrafish caudal fin, an optically accessible organ formed of repeating bony ray skeletal units, to determine the cellular relationship between nerves, bones and endothelium. In adult zebrafish, we establish the presence of somatosensory axons running through the inside of the bony fin rays, juxtaposed with osteoblasts on the inner hemiray surface. During development we show that the caudal fin progresses through sequential stages of endothelial plexus formation, bony ray addition, ray innervation and endothelial remodeling. Surprisingly, the initial stages of fin morphogenesis proceed normally in animals lacking either fin endothelium or somatosensory nerves. Instead, we find that sp7+ osteoblasts are required for endothelial remodeling and somatosensory axon innervation in the developing fin. Overall, this study demonstrates that the proximal neurovascular-bone relationship in the adult caudal fin is established during fin organogenesis and suggests that ray-associated osteoblasts pattern axons and endothelium.
Collapse
Affiliation(s)
- Rosalind G Bump
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Camille E A Goo
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Emma C Horton
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Jeffrey P Rasmussen
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
35
|
Abstract
Fracture healing is a complex, multistep process that is highly sensitive to mechanical signaling. To optimize repair, surgeons prescribe immediate weight-bearing as-tolerated within 24 hours after surgical fixation; however, this recommendation is based on anecdotal evidence and assessment of bulk healing outcomes (e.g., callus size, bone volume, etc.). Given challenges in accurately characterizing the mechanical environment and the ever-changing properties of the regenerate, the principles governing mechanical regulation of repair, including their cell and molecular basis, are not yet well defined. However, the use of mechanobiological rodent models, and their relatively large genetic toolbox, combined with recent advances in imaging approaches and single-cell analyses is improving our understanding of the bone microenvironment in response to loading. This review describes the identification and characterization of distinct cell populations involved in bone healing and highlights the most recent findings on mechanical regulation of bone homeostasis and repair with an emphasis on osteo-angio coupling. A discussion on aging and its impact on bone mechanoresponsiveness emphasizes the need for novel mechanotherapeutics that can re-sensitize skeletal stem and progenitor cells to physical rehabilitation protocols.
Collapse
Affiliation(s)
- Tareq Anani
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA
| | - Alesha B Castillo
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA; Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 11201, USA; Department of Veterans Affairs, New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010, USA.
| |
Collapse
|
36
|
Buettmann EG, Yoneda S, Hu P, McKenzie JA, Silva MJ. Postnatal Osterix but not DMP1 lineage cells significantly contribute to intramembranous ossification in three preclinical models of bone injury. Front Physiol 2022; 13:1083301. [PMID: 36685200 PMCID: PMC9846510 DOI: 10.3389/fphys.2022.1083301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/02/2022] [Indexed: 01/06/2023] Open
Abstract
Murine models of long-bone fracture, stress fracture, and cortical defect are used to discern the cellular and molecular mediators of intramembranous and endochondral bone healing. Previous work has shown that Osterix (Osx+) and Dentin Matrix Protein-1 (DMP1+) lineage cells and their progeny contribute to injury-induced woven bone formation during femoral fracture, ulnar stress fracture, and tibial cortical defect repair. However, the contribution of pre-existing versus newly-derived Osx+ and DMP1+ lineage cells in these murine models of bone injury is unclear. We addressed this knowledge gap by using male and female 12-week-old, tamoxifen-inducible Osx Cre_ERT2 and DMP1 Cre_ERT2 mice harboring the Ai9 TdTomato reporter allele. To trace pre-existing Osx+ and DMP1+ lineage cells, tamoxifen (TMX: 100 mg/kg gavage) was given in a pulse manner (three doses, 4 weeks before injury), while to label pre-existing and newly-derived lineage Osx+ and DMP1+ cells, TMX was first given 2 weeks before injury and continuously (twice weekly) throughout healing. TdTomato positive (TdT+) cell area and cell fraction were quantified from frozen histological sections of injured and uninjured contralateral samples at times corresponding with active woven bone formation in each model. We found that in uninjured cortical bone tissue, Osx Cre_ERT2 was more efficient than DMP1 Cre_ERT2 at labeling the periosteal and endosteal surfaces, as well as intracortical osteocytes. Pulse-labeling revealed that pre-existing Osx+ lineage and their progeny, but not pre-existing DMP1+ lineage cells and their progeny, significantly contributed to woven bone formation in all three injury models. In particular, these pre-existing Osx+ lineage cells mainly lined new woven bone surfaces and became embedded as osteocytes. In contrast, with continuous dosing, both Osx+ and DMP1+ lineage cells and their progeny contributed to intramembranous woven bone formation, with higher TdT+ tissue area and cell fraction in Osx+ lineage versus DMP1+ lineage calluses (femoral fracture and ulnar stress fracture). Similarly, Osx+ and DMP1+ lineage cells and their progeny significantly contributed to endochondral callus regions with continuous dosing only, with higher TdT+ chondrocyte fraction in Osx+ versus DMP1+ cell lineages. In summary, pre-existing Osx+ but not DMP1+ lineage cells and their progeny make up a significant amount of woven bone cells (particularly osteocytes) across three preclinical models of bone injury. Therefore, Osx+ cell lineage modulation may prove to be an effective therapy to enhance bone regeneration.
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States.,Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Pei Hu
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
37
|
Valdés-Fernández J, López-Martínez T, Ripalda-Cemboráin P, Calvo IA, Sáez B, Romero-Torrecilla JA, Aldazabal J, Muiños-López E, Montiel V, Orbe J, Rodríguez JA, Páramo JA, Prósper F, Granero-Moltó F. Molecular and Cellular Mechanisms of Delayed Fracture Healing in Mmp10 (Stromelysin 2) Knockout Mice. J Bone Miner Res 2021; 36:2203-2213. [PMID: 34173256 DOI: 10.1002/jbmr.4403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 11/08/2022]
Abstract
The remodeling of the extracellular matrix is a central function in endochondral ossification and bone homeostasis. During secondary fracture healing, vascular invasion and bone growth requires the removal of the cartilage intermediate and the coordinate action of the collagenase matrix metalloproteinase (MMP)-13, produced by hypertrophic chondrocytes, and the gelatinase MMP-9, produced by cells of hematopoietic lineage. Interfering with these MMP activities results in impaired fracture healing characterized by cartilage accumulation and delayed vascularization. MMP-10, Stromelysin 2, a matrix metalloproteinase with high homology to MMP-3 (Stromelysin 1), presents a wide range of putative substrates identified in vitro, but its targets and functions in vivo and especially during fracture healing and bone homeostasis are not well defined. Here, we investigated the role of MMP-10 through bone regeneration in C57BL/6 mice. During secondary fracture healing, MMP-10 is expressed by hematopoietic cells and its maximum expression peak is associated with cartilage resorption at 14 days post fracture (dpf). In accordance with this expression pattern, when Mmp10 is globally silenced, we observed an impaired fracture-healing phenotype at 14 dpf, characterized by delayed cartilage resorption and TRAP-positive cell accumulation. This phenotype can be rescued by a non-competitive transplant of wild-type bone marrow, indicating that MMP-10 functions are required only in cells of hematopoietic linage. In addition, we found that this phenotype is a consequence of reduced gelatinase activity and the lack of proMMP-9 processing in macrophages. Our data provide evidence of the in vivo function of MMP-10 during endochondral ossification and defines the macrophages as the lead cell population in cartilage removal and vascular invasion. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | | | - Purificación Ripalda-Cemboráin
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Isabel A Calvo
- Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Borja Sáez
- Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | | | - Javier Aldazabal
- Tissue Engineering Group, TECNUN-Universidad de Navarra, San Sebastián, Spain
| | | | - Verónica Montiel
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Josune Orbe
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain
| | - José Antonio Rodríguez
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain
| | - José Antonio Páramo
- Atherotrombosis, Cardiovascular Disease Program, CIMA, Instituto de Investigación Sanitaria de Navarra (IdiSNA), CIBERCV, Pamplona, Spain.,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prósper
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Hematology-Oncology Program, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Froilán Granero-Moltó
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain.,Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
38
|
Zhang X, Wang H, Hao Z. A numerical bone regeneration model incorporating angiogenesis, considering oxygen-induced secretion of vascular endothelial growth factor and vascular remodeling. J Biomech 2021; 127:110656. [PMID: 34416529 DOI: 10.1016/j.jbiomech.2021.110656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 05/06/2021] [Accepted: 07/25/2021] [Indexed: 02/07/2023]
Abstract
Angiogenesis is considered playing an important role in bone regeneration. Studies have shown that angiogenesis is affected by biological factors, oxygen tension, and blood flow. In this paper, we propose a bone regeneration model with angiogenesis based on the theories of mechanobiology regulation, vascular network modeling, oxygen-induced secretion of vascular endothelial growth factor (VEGF), and vascular remodeling. The results showed that this model can describe the distribution and concentration of vascular endothelial growth factor induced by oxygen tension during bone regeneration, the growth and remodeling of vascular tissue under the influence of vascular endothelial growth factor and mechanical loading, and the correspondence between vascular tissue and bone regeneration.
Collapse
Affiliation(s)
- Xuanbin Zhang
- The State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Haosen Wang
- The State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Zhixiu Hao
- The State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
39
|
Graham SM, Maqungo S, Laubscher M, Ferreira N, Held M, Harrison WJ, Simpson AH, MacPherson P, Lalloo DG. Fracture Healing in Patients With HIV in South Africa: A Prospective Cohort Study. J Acquir Immune Defic Syndr 2021; 87:1214-1220. [PMID: 33990496 PMCID: PMC8263144 DOI: 10.1097/qai.0000000000002720] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/19/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND HIV reduces bone mineral density, mineralization, and turnover and may impair fracture healing. SETTING This prospective cohort study in South Africa investigated whether HIV infection was associated with impaired fracture healing after trauma. METHODS All adults with acute tibia and femur fractures who underwent intermedullary (IM) nailing for fracture fixation between September 2017 and December 2018, at 2 tertiary hospitals, were followed up for a minimum of 12 months postoperatively. The primary outcome was delayed bone union at 6 months (defined by the radiological union scoring system for the tibia score <9), and the secondary outcome was nonunion (defined as radiological union scoring system for the tibia score <9) at 9 months. Multivariable logistic regression models were constructed to investigate the associations between HIV status and impaired fracture healing. RESULTS In total, 358 participants, who underwent 395 IM nailings, were enrolled in the study and followed up for 12 months. Seventy-one of the 358 (19.8%) participants were HIV-positive [83/395 (21%) IM nailings]. HIV was not associated with delayed fracture healing after IM nailing of the tibia or femur (multivariable odds ratio: 1.06; 95% confidence interval: 0.50 to 2.22). HIV-positive participants had a statistically significant lower odds ratio of nonunion compared with HIV-negative participants (multivariable odds ratio: 0.17; 95% confidence interval: 0.01 to 0.92). CONCLUSIONS Fractures sustained in HIV-positive individuals can undergo surgical fixation as effectively as those in HIV-negative individuals, with no increased risk of delayed union or nonunion.
Collapse
Affiliation(s)
- Simon M. Graham
- Institute of Population Health Data Science, University of Liverpool, Liverpool, United Kingdom;
- Department of Orthopaedic and Trauma Surgery, Liverpool University Teaching Hospital Trust, Liverpool, United Kingdom;
- Division of Orthopaedic Surgery, Groote Schuur Hospital, Cape Town, South Africa;
| | - Sithombo Maqungo
- Division of Orthopaedic Surgery, Groote Schuur Hospital, Cape Town, South Africa;
- Division of Global Surgery, Orthopaedic Research Unit (ORU), University of Cape Town, Cape Town, South Africa;
| | - Maritz Laubscher
- Division of Orthopaedic Surgery, Groote Schuur Hospital, Cape Town, South Africa;
- Division of Global Surgery, Orthopaedic Research Unit (ORU), University of Cape Town, Cape Town, South Africa;
| | - Nando Ferreira
- Division of Orthopaedic Surgery, Stellenbosch University, Cape Town, South Africa;
| | - Michael Held
- Division of Orthopaedic Surgery, Groote Schuur Hospital, Cape Town, South Africa;
- Division of Global Surgery, Orthopaedic Research Unit (ORU), University of Cape Town, Cape Town, South Africa;
| | - William J. Harrison
- Institute of Population Health Data Science, University of Liverpool, Liverpool, United Kingdom;
- Department of Orthopaedic and Trauma Surgery, Countess of Chester Hospital, Chester, United Kingdom;
| | | | - Peter MacPherson
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom; and
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.
| | - David G. Lalloo
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.
| |
Collapse
|
40
|
Systemic Administration of PTH Supports Vascularization in Segmental Bone Defects Filled with Ceramic-Based Bone Graft Substitute. Cells 2021; 10:cells10082058. [PMID: 34440827 PMCID: PMC8392660 DOI: 10.3390/cells10082058] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/29/2021] [Accepted: 08/07/2021] [Indexed: 12/19/2022] Open
Abstract
Non-unions continue to present a challenge to trauma surgeons, as current treatment options are limited, duration of treatment is long, and the outcome often unsatisfactory. Additionally, standard treatment with autologous bone grafts is associated with comorbidity at the donor site. Therefore, alternatives to autologous bone grafts and further therapeutic strategies to improve on the outcome and reduce cost for care providers are desirable. In this study in Sprague–Dawley rats we employed a recently established sequential defect model, which provides a platform to test new potential therapeutic strategies on non-unions while gaining mechanistic insight into their actions. The effects of a combinatorial treatment of a bone graft substitute (HACaS+G) implantation and systemic PTH administration was assessed by µ-CT, histological analysis, and bio-mechanical testing and compared to monotreatment and controls. Although neither PTH alone nor the combination of a bone graft substitute and PTH led to the formation of a stable union, our data demonstrate a clear osteoinductive and osteoconductive effect of the bone graft substitute. Additionally, PTH administration was shown to induce vascularization, both as a single adjuvant treatment and in combination with the bone graft substitute. Thus, systemic PTH administration is a potential synergistic co-treatment to bone graft substitutes.
Collapse
|
41
|
A Chemotactic Functional Scaffold with VEGF-Releasing Peptide Amphiphiles Facilitates Bone Regeneration by BMP-2 in a Large-Scale Rodent Cranial Defect Model. Plast Reconstr Surg 2021; 147:386-397. [PMID: 33235044 DOI: 10.1097/prs.0000000000007551] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Current common techniques for repairing calvarial defects by autologous bone grafting and alloplastic implants have significant limitations. In this study, the authors investigated a novel alternative approach to bone repair based on peptide amphiphile nanofiber gels that are engineered to control the release of vascular endothelial growth factor (VEGF) to recruit circulating stem cells to a site of bone regeneration and facilitate bone healing by bone morphogenetic protein-2 (BMP-2). METHODS VEGF release kinetics from peptide amphiphile gels were evaluated. Chemotactic functional scaffolds were fabricated by combining collagen sponges with peptide amphiphile gels containing VEGF. The in vitro and in vivo chemotactic activities of the scaffolds were evaluated by measuring mesenchymal stem cell migration, and angiogenic capability of the scaffolds was also evaluated. Large-scale rodent cranial bone defects were created to evaluate bone regeneration after implanting the scaffolds and other control materials. RESULTS VEGF was released from peptide amphiphile in a controlled-release manner. In vitro migration of mesenchymal stem cells was significantly greater when exposed to chemotactic functional scaffolds compared to control scaffolds. In vivo chemotaxis was evidenced by migration of tracer-labeled mesenchymal stem cells to the chemotactic functional scaffolds. Chemotactic functional scaffolds showed significantly increased angiogenesis in vivo. Successful bone regeneration was noted in the defects treated with chemotactic functional scaffolds and BMP-2. CONCLUSIONS The authors' observations suggest that this bioengineered construct successfully acts as a chemoattractant for circulating mesenchymal stem cells because of controlled release of VEGF from the peptide amphiphile gels. The chemotactic functional scaffolds may play a role in the future design of clinically relevant bone graft substitutes for large-scale bone defects.
Collapse
|
42
|
The Cellular Choreography of Osteoblast Angiotropism in Bone Development and Homeostasis. Int J Mol Sci 2021; 22:ijms22147253. [PMID: 34298886 PMCID: PMC8305002 DOI: 10.3390/ijms22147253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
Interaction between endothelial cells and osteoblasts is essential for bone development and homeostasis. This process is mediated in large part by osteoblast angiotropism, the migration of osteoblasts alongside blood vessels, which is crucial for the homing of osteoblasts to sites of bone formation during embryogenesis and in mature bones during remodeling and repair. Specialized bone endothelial cells that form "type H" capillaries have emerged as key interaction partners of osteoblasts, regulating osteoblast differentiation and maturation and ensuring their migration towards newly forming trabecular bone areas. Recent revolutions in high-resolution imaging methodologies for bone as well as single cell and RNA sequencing technologies have enabled the identification of some of the signaling pathways and molecular interactions that underpin this regulatory relationship. Similarly, the intercellular cross talk between endothelial cells and entombed osteocytes that is essential for bone formation, repair, and maintenance are beginning to be uncovered. This is a relatively new area of research that has, until recently, been hampered by a lack of appropriate analysis tools. Now that these tools are available, greater understanding of the molecular relationships between these key cell types is expected to facilitate identification of new drug targets for diseases of bone formation and remodeling.
Collapse
|
43
|
Wang C, Ying J, Nie X, Zhou T, Xiao D, Swarnkar G, Abu-Amer Y, Guan J, Shen J. Targeting angiogenesis for fracture nonunion treatment in inflammatory disease. Bone Res 2021; 9:29. [PMID: 34099632 PMCID: PMC8184936 DOI: 10.1038/s41413-021-00150-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 02/05/2023] Open
Abstract
Atrophic fracture nonunion poses a significant clinical problem with limited therapeutic interventions. In this study, we developed a unique nonunion model with high clinical relevance using serum transfer-induced rheumatoid arthritis (RA). Arthritic mice displayed fracture nonunion with the absence of fracture callus, diminished angiogenesis and fibrotic scar tissue formation leading to the failure of biomechanical properties, representing the major manifestations of atrophic nonunion in the clinic. Mechanistically, we demonstrated that the angiogenesis defect observed in RA mice was due to the downregulation of SPP1 and CXCL12 in chondrocytes, as evidenced by the restoration of angiogenesis upon SPP1 and CXCL12 treatment in vitro. In this regard, we developed a biodegradable scaffold loaded with SPP1 and CXCL12, which displayed a beneficial effect on angiogenesis and fracture repair in mice despite the presence of inflammation. Hence, these findings strongly suggest that the sustained release of SPP1 and CXCL12 represents an effective therapeutic approach to treat impaired angiogenesis and fracture nonunion under inflammatory conditions.
Collapse
Affiliation(s)
- Cuicui Wang
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| | - Jun Ying
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA ,grid.417400.60000 0004 1799 0055Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China ,grid.417400.60000 0004 1799 0055Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolei Nie
- grid.4367.60000 0001 2355 7002Department of Mechanical Engineering & Materials Science, School of Engineering, Washington University, St. Louis, MO USA
| | - Tianhong Zhou
- grid.4367.60000 0001 2355 7002Department of Mechanical Engineering & Materials Science, School of Engineering, Washington University, St. Louis, MO USA
| | - Ding Xiao
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| | - Gaurav Swarnkar
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| | - Yousef Abu-Amer
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA ,grid.415840.c0000 0004 0449 6533Shriners Hospital for Children, St. Louis, MO USA
| | - Jianjun Guan
- grid.4367.60000 0001 2355 7002Department of Mechanical Engineering & Materials Science, School of Engineering, Washington University, St. Louis, MO USA
| | - Jie Shen
- grid.4367.60000 0001 2355 7002Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO USA
| |
Collapse
|
44
|
Kulanthaivel S, Agarwal T, Sharan Rathnam VS, Pal K, Banerjee I. Cobalt doped nano-hydroxyapatite incorporated gum tragacanth-alginate beads as angiogenic-osteogenic cell encapsulation system for mesenchymal stem cell based bone tissue engineering. Int J Biol Macromol 2021; 179:101-115. [PMID: 33621571 DOI: 10.1016/j.ijbiomac.2021.02.136] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
Angiogenic-osteogenic cell encapsulation system is a technical need for human mesenchymal stem cell (hMSC)-based bone tissue engineering (BTE). Here, we have developed a highly efficient hMSC encapsulation system by incorporating bivalent cobalt doped nano-hydroxyapatite (HAN) and gum tragacanth (GT) as angiogenic-osteogenic components into the calcium alginate (CA) beads. Physico-chemical characterizations revealed that the swelling and degradation of HAN incorporated CA-GT beads (GT-HAN) were 1.34 folds and 2 folds higher than calcium alginate (CA) beads. Furthermore, the diffusion coefficient of solute molecule was found 2.5-fold higher in GT-HAN with respect to CA bead. It is observed that GT-HAN supports the long-term viability of encapsulated hMSC and causes 50% less production of reactive oxygen species (ROS) in comparison to CA beads. The expression of osteogenic differentiation markers was found 1.5-2.5 folds higher in the case of GT-HAN in comparison to CA. A similar trend was observed for hypoxia inducible factor 1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF). The soluble secretome from hMSC encapsulated in the GT-HAN induced proliferation of endothelial cells and supported tube formation (2.5-fold higher than CA beads). These results corroborated that GT-HAN could be used as an angiogenic-osteogenic cell encapsulation matrix for hMSC encapsulation and BTE application.
Collapse
Affiliation(s)
- Senthilguru Kulanthaivel
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - V S Sharan Rathnam
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Kunal Pal
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Indranil Banerjee
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Rajasthan 342037, India.
| |
Collapse
|
45
|
Abstract
Bone is a highly vascularized tissue. However, despite the importance of appropriate circulation for bone health, regulation of bone blood flow remains poorly understood. Invasive animal studies suggest that sympathetic activity plays an important role in bone flow control. However, it remains unknown if bone vasculature evidences robust vasoconstriction in response to sympathoexcitatory stimuli. Here, we characterized bone blood flow in young healthy individuals [n = 13, (four females)] in response to isometric handgrip exercise (IHE) and cold pressor test (CPT). These provide a strong stimulus for active vasoconstriction in the inactive muscle, and perhaps also in the bone. During sustained IHE to fatigue and CPT, we measured blood pressure, whole leg blood flow, and tibial perfusion using near-infrared spectroscopy. Tibia perfusion was determined as oxy- and deoxyhemoglobin. For both stimuli, tibial metabolism remained constant (i.e., no change in deoxyhemoglobin) and thus tibial arterial perfusion was represented by oxyhemoglobin. During IHE, oxyhemoglobin declined (beginning -0.20 ± 1.04 μM; end -1.13 ± 3.71 μM, both P < 0.01) slower than whole leg blood flow (beginning -0.85 ± 1.02 cm/s; end -2.72 ± 1.64 cm/s, both P < 0.01). However, during CPT, both oxyhemoglobin (beginning -0.46 ± 1.43 μM; end -0.60 ± 1.59 μM, both P < 0.01) and whole leg blood flow (beginning -1.52 ± 1.63 cm/s; end -0.69 ± 1.51 cm/s, both P < 0.01) declined with a similar timecourse, even though the magnitudes of decline were smaller than during IHE. These responses are likely due to the different timecourses of sympathetically mediated vasoconstriction in bone and muscle. These results indicate that sympathetic innervation of the bone vasculature serves a functional role in the control of flow in young healthy individuals.NEW & NOTEWORTHY The current study is the first one to noninvasively investigate control of bone blood perfusion in vivo in humans, on a moment-by-moment basis. Our results indicate that tibial bone vasculature demonstrates active vasoconstriction in response to sympathoexcitatory stimuli in young healthy individuals. Compared with whole leg vasculature, bone vasoconstrictor response seems to be smaller, delayed, and more variable.
Collapse
Affiliation(s)
- Adina E Draghici
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts.,Cardiovascular Research Laboratory, Spaulding Hospital Cambridge, Cambridge, Massachusetts
| | - J Andrew Taylor
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts.,Cardiovascular Research Laboratory, Spaulding Hospital Cambridge, Cambridge, Massachusetts
| |
Collapse
|
46
|
Duan ZW, Lu H. Effect of Mechanical Strain on Cells Involved in Fracture Healing. Orthop Surg 2021; 13:369-375. [PMID: 33496077 PMCID: PMC7957396 DOI: 10.1111/os.12885] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/08/2020] [Accepted: 10/26/2020] [Indexed: 12/30/2022] Open
Abstract
Secondary fracture healing is a complex multi‐stage process in which the mechanical environment plays a key role. The use of an appropriate mechanical stimulation such as strain is conducive to tissue formation between fracture ends, thus aiding the healing process. However, if the strain is too large or too small, the biological behavior of the cells involved in bone healing will be affected, resulting in non‐union or delayed healing. In this review, we summarize the current state of knowledge regarding the effect of strain on cells that play a role in the fracture‐healing process. Overall, the related literature suggests that selection of an adequate strain promotes fracture healing through the stimulation of angiogenesis and osteogenesis, along with inhibition of osteoclast differentiation and bone resorption. However, standardized methods for the application of mechanical stimulation are lacking, and a unified consensus on the mechanism by which strain promotes cell differentiation has not yet been reached. These issues, therefore, deserve further investigation.
Collapse
Affiliation(s)
- Zheng-Wei Duan
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Lu
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Oda T, Niikura T, Fukui T, Oe K, Kuroiwa Y, Kumabe Y, Sawauchi K, Yoshikawa R, Mifune Y, Hayashi S, Matsumoto T, Matsushita T, Kawamoto T, Sakai Y, Akisue T, Kuroda R. Transcutaneous CO 2 application accelerates fracture repair in streptozotocin-induced type I diabetic rats. BMJ Open Diabetes Res Care 2020; 8:8/2/e001129. [PMID: 33323458 PMCID: PMC7745327 DOI: 10.1136/bmjdrc-2019-001129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 10/29/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Diabetes mellitus (DM) negatively affects fracture repair by inhibiting endochondral ossification, chondrogenesis, callus formation, and angiogenesis. We previously reported that transcutaneous CO2 application accelerates fracture repair by promoting endochondral ossification and angiogenesis. The present study aimed to determine whether CO2 treatment would promote fracture repair in cases with type I DM. RESEARCH DESIGN AND METHODS A closed femoral shaft fracture was induced in female rats with streptozotocin-induced type I DM. CO2 treatment was performed five times a week for the CO2 group. Sham treatment, where CO2 was replaced with air, was performed for the control group. Radiographic, histologic, genetic, and biomechanical measurements were taken at several time points. RESULTS Radiographic assessment demonstrated that fracture repair was induced in the CO2 group. Histologically, accelerated endochondral ossification and capillary formation were observed in the CO2 group. Immunohistochemical assessment indicated that early postfracture proliferation of chondrocytes in callus was enhanced in the CO2 group. Genetic assessment results suggested that cartilage and bone formation, angiogenesis, and vasodilation were upregulated in the CO2 group. Biomechanical assessment revealed enhanced mechanical strength in the CO2 group. CONCLUSIONS Our findings suggest that CO2 treatment accelerates fracture repair in type I DM rats. CO2 treatment could be an effective strategy for delayed fracture repair due to DM.
Collapse
Affiliation(s)
- Takahiro Oda
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Takahiro Niikura
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Tomoaki Fukui
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Keisuke Oe
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Yu Kuroiwa
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Yohei Kumabe
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Kenichi Sawauchi
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Ryo Yoshikawa
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Yutaka Mifune
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Shinya Hayashi
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Tomoyuki Matsumoto
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Takehiko Matsushita
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Teruya Kawamoto
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Yoshitada Sakai
- Division of Rehabilitation Medicine, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| | - Toshihiro Akisue
- Department of Rehabilitation Science, Kobe University Faculty of Health Sciences and Graduate School of Medicine Faculty of Health Sciences, Kobe, Hyogo, Japan
| | - Ryosuke Kuroda
- Orthopaedic Surgery, Kobe University Graduate School of Medicine School of Medicine, Kobe, Hyogo, Japan
| |
Collapse
|
48
|
Stucker S, Chen J, Watt FE, Kusumbe AP. Bone Angiogenesis and Vascular Niche Remodeling in Stress, Aging, and Diseases. Front Cell Dev Biol 2020; 8:602269. [PMID: 33324652 PMCID: PMC7726257 DOI: 10.3389/fcell.2020.602269] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/05/2020] [Indexed: 02/05/2023] Open
Abstract
The bone marrow (BM) vascular niche microenvironments harbor stem and progenitor cells of various lineages. Bone angiogenesis is distinct and involves tissue-specific signals. The nurturing vascular niches in the BM are complex and heterogenous consisting of distinct vascular and perivascular cell types that provide crucial signals for the maintenance of stem and progenitor cells. Growing evidence suggests that the BM niche is highly sensitive to stress. Aging, inflammation and other stress factors induce changes in BM niche cells and their crosstalk with tissue cells leading to perturbed hematopoiesis, bone angiogenesis and bone formation. Defining vascular niche remodeling under stress conditions will improve our understanding of the BM vascular niche and its role in homeostasis and disease. Therefore, this review provides an overview of the current understanding of the BM vascular niches for hematopoietic stem cells and their malfunction during aging, bone loss diseases, arthritis and metastasis.
Collapse
Affiliation(s)
- Sina Stucker
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Junyu Chen
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fiona E. Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Assefa F, Lim J, Kim JA, Ihn HJ, Lim S, Nam SH, Bae YC, Park EK. Secretoneurin, a Neuropeptide, Enhances Bone Regeneration in a Mouse Calvarial Bone Defect Model. Tissue Eng Regen Med 2020; 18:315-324. [PMID: 33145742 DOI: 10.1007/s13770-020-00304-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND This study investigates the effects of a neuropeptide, secretoneurin (SN), on bone regeneration in an experimental mouse model. METHODS The effects of SN on cell proliferation, osteoblast marker genes expression, and mineralization were evaluated using the CCK-8 assay, quantitative reverse transcriptase polymerase chain reaction (RT-PCR), and alizarin red S staining, respectively. To examine the effects of SN on bone regeneration in vivo, bone defects were created in the calvaria of ICR mice, and 0.5 or 1 µg/ml SN was applied. New bone formation was analyzed by micro-computed tomography (micro-CT) and histology. New blood vessel formation was assessed by CD34 immunohistochemistry. RESULTS SN had no significant effect on proliferation and mineralization of MC3T3-E1 cells. However, SN partially induced the gene expression of osteoblast differentiation markers such as runt-related transcription factor 2, alkaline phosphatase, collagen type I alpha 1, and osteopontin. A significant increase of bone regeneration was observed in SN treated calvarial defects. The bone volume (BV), BV/tissue volume, trabecular thickness and trabecular number values were significantly increased in the collagen sponge plus 0.5 or 1 µg/ml SN group (p < 0.01) compared with the control group. Histologic analysis also revealed increased new bone formation in the SN-treated groups. Immunohistochemical staining of CD34 showed that the SN-treated groups contained more blood vessels compared with control in the calvarial defect area. CONCLUSION SN increases new bone and blood vessel formation in a calvarial defect site. This study suggests that SN may enhance new bone formation through its potent angiogenic activity.
Collapse
Affiliation(s)
- Freshet Assefa
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Jiwon Lim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Ju-Ang Kim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Hye Jung Ihn
- Cell & Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Soomin Lim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Sang-Hyeon Nam
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea
| | - Yong Chul Bae
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, South Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 41940, Republic of Korea.
| |
Collapse
|
50
|
Huang S, Jin M, Su N, Chen L. New insights on the reparative cells in bone regeneration and repair. Biol Rev Camb Philos Soc 2020; 96:357-375. [PMID: 33051970 DOI: 10.1111/brv.12659] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Bone possesses a remarkable repair capacity to regenerate completely without scar tissue formation. This unique characteristic, expressed during bone development, maintenance and injury (fracture) healing, is performed by the reparative cells including skeletal stem cells (SSCs) and their descendants. However, the identity and functional roles of SSCs remain controversial due to technological difficulties and the heterogeneity and plasticity of SSCs. Moreover, for many years, there has been a biased view that bone marrow is the main cell source for bone repair. Together, these limitations have greatly hampered our understanding of these important cell populations and their potential applications in the treatment of fractures and skeletal diseases. Here, we reanalyse and summarize current understanding of the reparative cells in bone regeneration and repair and outline recent progress in this area, with a particular emphasis on the temporal and spatial process of fracture healing, the sources of reparative cells, an updated definition of SSCs, and markers of skeletal stem/progenitor cells contributing to the repair of craniofacial and long bones, as well as the debate between SSCs and pericytes. Finally, we also discuss the existing problems, emerging novel technologies and future research directions in this field.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| |
Collapse
|