1
|
Özkan ÖC, Kurdal DP, Yılmaz B, Tutcu HK, Somuncu ÖS, Yücel IA, Savaşır E, Midi A. Comparison of the Effects of Microfracture, Soft Callus Implantation, and Matrix-Supported Chondrocyte Implantation in an Experimental Osteochondral Defect Model in Rats. Niger J Clin Pract 2024; 27:1154-1163. [DOI: 10.4103/njcp.njcp_134_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/26/2024] [Indexed: 01/03/2025]
Abstract
Background:
The treatment of cartilage defects remains challenging due to the avascular nature of cartilage.
Aim:
This study investigates the therapeutic effect of soft callus in osteochondral defects and explores the ability of multipotent and pluripotent cells within the callus to form fibrous or hyaline cartilage in the defective area.
Methods:
Twenty-one rats were divided into three equal groups: Group 1 received only microfracture (MF), group 2 received microfracture with autologous chondrocyte implantation (MF+ACI), and group 3 received microfracture with soft callus implantation (MF+SCI). All rats underwent diaphyseal fracture in their left tibias, which was stabilized with a Kirshner wire. One week later, osteochondral defects were created in the right knees of all rats. For group 1, microfracture alone was applied to initiate healing in the defects. In group 2, heterologous chondrocytes, previously harvested from the lateral condyle of a rat’s left femur and cultivated in a laboratory environment, were implanted into the microfracture site. In group 3, soft callus tissue obtained from the left tibial fracture was compressed and implanted into the defective area. All groups were sacrificed at the 6th week, and the healing status of the osteochondral defect areas was histopathologically evaluated.
Results:
Macroscopic examination at the end of the study revealed comparable ICRS-1 scores for MF+ACI (group 2) (11.28 ± 1.25) and MF+SCI (group 3) (11.14 ± 0.37), while MF alone (group 1) (4.28 ± 1.25) showed significantly lower results. Microscopic examination yielded similar outcomes. Regarding histological scores, ICRS-2 scores for MF (group 1) (35.30 ± 1.13), MF+ACI (group 2) (47.09 ± 1.63), and MF+SCI (group 3) (43.97 ± 1.49) were statistically significantly lower.
Conclusion:
Defects treated with soft callus implantation demonstrated comparable outcomes to the widely used and gold-standard autologous chondrocyte implantation. When compared to microfracture alone, better macroscopic and microscopic results were achieved with soft callus implantation.
Collapse
Affiliation(s)
- ÖC Özkan
- Department of Orthopedic, Fatih Sultan Mehmet, Training and Research Hospital, Istanbul, Turkey
| | - DP Kurdal
- Department of Orthopedic, Fatih Sultan Mehmet, Training and Research Hospital, Istanbul, Turkey
| | - B Yılmaz
- Department of Orthopedic, Fatih Sultan Mehmet, Training and Research Hospital, Istanbul, Turkey
| | - HK Tutcu
- Department of Orthopedic, Fatih Sultan Mehmet, Training and Research Hospital, Istanbul, Turkey
| | - ÖS Somuncu
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, United States
| | - IA Yücel
- Department of Orthopedic, Fatih Sultan Mehmet, Training and Research Hospital, Istanbul, Turkey
| | - E Savaşır
- Department of Medicine, University of Bahcesehir, Istanbul, Turkey
| | - A Midi
- Department of Pathology, School of Medicine, University of Altınbaş, Istanbul, Turkey
| |
Collapse
|
2
|
Jiang W, Caruana DL, Back J, Lee FY. Unique Spatial Transcriptomic Profiling of the Murine Femoral Fracture Callus: A Preliminary Report. Cells 2024; 13:522. [PMID: 38534368 PMCID: PMC10969736 DOI: 10.3390/cells13060522] [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: 02/03/2024] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Fracture callus formation is a dynamic stage of bone activity and repair with precise, spatially localized gene expression. Metastatic breast cancer impairs fracture healing by disrupting bone homeostasis and imparting an altered genomic profile. Previous sequencing techniques such as single-cell RNA and in situ hybridization are limited by missing spatial context and low throughput, respectively. We present a preliminary approach using the Visium CytAssist spatial transcriptomics platform to provide the first spatially intact characterization of genetic expression changes within an orthopedic model of impaired fracture healing. Tissue slides prepared from BALB/c mice with or without MDA-MB-231 metastatic breast cancer cells were used. Both unsupervised clustering and histology-based annotations were performed to identify the hard callus, soft callus, and interzone for differential gene expression between the wild-type and pathological fracture model. The spatial transcriptomics platform successfully localized validated genes of the hard (Dmp1, Sost) and soft callus (Acan, Col2a1). The fibrous interzone was identified as a region of extensive genomic heterogeneity. MDA-MB-231 samples demonstrated downregulation of the critical bone matrix and structural regulators that may explain the weakened bone structure of pathological fractures. Spatial transcriptomics may represent a valuable tool in orthopedic research by providing temporal and spatial context.
Collapse
Affiliation(s)
| | | | | | - Francis Y. Lee
- Department of Orthopaedics & Rehabilitation, Yale School of Medicine, 47 College Place, New Haven, CT 06510, USA
| |
Collapse
|
3
|
Tong L, Yang Z, Dai W, Sun Z, Yang J, Xue Q, Li Y. Experimental study on determining the degree of bone healing by wall thickness ratio analysis. J Orthop Surg Res 2024; 19:79. [PMID: 38243260 PMCID: PMC10799492 DOI: 10.1186/s13018-024-04565-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024] Open
Abstract
To verify the reliability and accuracy of wall thickness ratio analysis to determine the degree of bone healing, fracture models were established with 6 beagles. X-ray, micro-CT, and CT scans were performed at 24 weeks. The healthy side and the affected side were used to simulate the three-dimensional geometric model after internal fixation, and the mesh was divided. The mean and median CT wall thickness values were obtained through the wall thickness analysis. X-ray, CT, micro-CT, and gross appearance were used to determine the degree of bone healing, which was compared with wall thickness analysis. There was a positive correlation between the average CT value and the median wall thickness. The correlation coefficient analysis of the median wall thickness ratio (R2) and healing index ratio (R3) showed a positive correlation. The results of the wall thickness ratio (R2) and the healing index ratio (R3) were used to determine bone healing, and the results were consistent with the results of the actual mechanical test and image analysis. The results of wall thickness ratio analysis were significantly correlated with the degree of bone healing. This method is simple, rapid, and practical to analyze and judge the degree of bone healing.
Collapse
Affiliation(s)
- Liangcheng Tong
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Zhiwei Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Wei Dai
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Zhongyang Sun
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Junsheng Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Qing Xue
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China
| | - Ying Li
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Anhui Medical University, No. 1 Malu Road, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
4
|
Palomino Lago E, Baird A, Blott SC, McPhail RE, Ross AC, Durward-Akhurst SA, Guest DJ. A Functional Single-Nucleotide Polymorphism Upstream of the Collagen Type III Gene Is Associated with Catastrophic Fracture Risk in Thoroughbred Horses. Animals (Basel) 2023; 14:116. [PMID: 38200847 PMCID: PMC10778232 DOI: 10.3390/ani14010116] [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: 11/16/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Fractures caused by bone overloading are a leading cause of euthanasia in Thoroughbred racehorses. The risk of fatal fracture has been shown to be influenced by both environmental and genetic factors but, to date, no specific genetic mechanisms underpinning fractures have been identified. In this study, we utilised a genome-wide polygenic risk score to establish an in vitro cell system to study bone gene regulation in horses at high and low genetic risk of fracture. Candidate gene expression analysis revealed differential expression of COL3A1 and STAT1 genes in osteoblasts derived from high- and low-risk horses. Whole-genome sequencing of two fracture cases and two control horses revealed a single-nucleotide polymorphism (SNP) upstream of COL3A1 that was confirmed in a larger cohort to be significantly associated with fractures. Bioinformatics tools predicted that this SNP may impact the binding of the transcription factor SOX11. Gene modulation demonstrated SOX11 is upstream of COL3A1, and the region binds to nuclear proteins. Furthermore, luciferase assays demonstrated that the region containing the SNP has promoter activity. However, the specific effect of the SNP depends on the broader genetic background of the cells and suggests other factors may also be involved in regulating COL3A1 expression. In conclusion, we have identified a novel SNP that is significantly associated with fracture risk and provide new insights into the regulation of the COL3A1 gene.
Collapse
Affiliation(s)
- Esther Palomino Lago
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, UK; (E.P.L.); (A.C.R.)
| | - Arabella Baird
- Animal Health Trust, Lanwades Park, Kentford, Newmarket CB8 7UU, UK
| | - Sarah C. Blott
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham LE12 5RD, UK;
| | - Rhona E. McPhail
- Animal Health Trust, Lanwades Park, Kentford, Newmarket CB8 7UU, UK
| | - Amy C. Ross
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, UK; (E.P.L.); (A.C.R.)
| | - Sian A. Durward-Akhurst
- Department of Veterinary Clinical Sciences, University of Minnesota, Saint Paul, MN 55108, USA;
| | - Deborah J. Guest
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, UK; (E.P.L.); (A.C.R.)
| |
Collapse
|
5
|
Li Y, Yang Z, Tong L, Yang J, Wang J, Wen Y. Wall thickness analysis method for judging the degree of lower extremity long bone healing. Sci Rep 2023; 13:20650. [PMID: 38001361 PMCID: PMC10673992 DOI: 10.1038/s41598-023-48212-3] [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: 11/19/2022] [Accepted: 11/23/2023] [Indexed: 11/26/2023] Open
Abstract
To evaluate the possibility of judging the degree of bone healing by wall thickness analysis provide reference for quantitative analysis of bone healing. Patients with lower limb fracture from April 2014 to October 2019 were recruited and divided into bone healing (group A), poor bone healing (group B), and nonunion (group C). Models were built in Mimics 20.0 with DICOM 3.0 data obtained from patient's CT. Three-dimensional geometric models of unaffected limb and affected limb after simulated removal of internal fixation were established, corresponding to basic phase and simulated phase, respectively. Wall thickness analysis was performed to obtain median wall thickness after meshing. R2 (median wall thickness ratio), R4 (CT value ratio), and R5 (healing index ratio) were obtained by calculating the ratio of each value in simulated phase to that in basic phase. Receiver operating characteristic curve analysis was used to evaluate the ability of Wall Thickness Analysis to indicate fracture healing. 112 CT scans of 79 patients were included in the study. The frequency of categorization in groups A, B, and C was 49, 37 and 26, respectively. The median R2 in groups A, B, and C was 0.91, 0.80, and 0.67, respectively (group A > group B > group C, all P < 0.05). The best cutoff point for R2 in predicting bone healing was 0.84, and predicting bone nonunion was 0.74. The Wall Thickness Analysis can be used to quantitatively evaluate fracture healing state, with median wall thickness ratio as a more intuitive and reliable judgment index.
Collapse
Affiliation(s)
- Ying Li
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
- Department of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Zhiwei Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Liangcheng Tong
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Junsheng Yang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Jianling Wang
- Department of Orthopedics, Air Force Hospital of Eastern Theater Command, Nanjing, Jiangsu, China
| | - Yaoke Wen
- School of Mechanical Engineering, Nanjing University of Science and Technology, No. 200 Xiaolingwei, Nanjing, 210094, Jiangsu, China.
| |
Collapse
|
6
|
Gu F, Zhang K, Zhu WA, Sui Z, Li J, Xie X, Yu T. Silicone rubber sealed channel induced self-healing of large bone defects: Where is the limit of self-healing of bone? J Orthop Translat 2023; 43:21-35. [PMID: 37965195 PMCID: PMC10641457 DOI: 10.1016/j.jot.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 11/16/2023] Open
Abstract
Background Large defects of long tubular bones due to severe trauma, bone tumor resection, or osteomyelitis debridement are challenging in orthopedics. Bone non-union and other complications often lead to serious consequences. At present, autologous bone graft is still the gold standard for the treatment of large bone defects. However, autologous bone graft sources are limited. Silicon rubber (SR) materials are widely used in biomedical fields, due to their safety and biocompatibility, and even shown to induce nerve regeneration. Materials and methods We extracted rat bone marrow mesenchymal stem cells (BMMSCs) in vitro and verified the biocompatibility of silicone rubber through cell experiments. Then we designed a rabbit radius critical sized bone defect model to verify the effect of silicone rubber sealed channel inducing bone repair in vivo. Results SR sealed channel could prevent the fibrous tissue from entering the fracture end and forming bone nonunion, thereby inducing self-healing of long tubular bone through endochondral osteogenesis. The hematoma tissue formed in the early stage was rich in osteogenesis and angiogenesis related proteins, and gradually turned into vascularization and endochondral osteogenesis, and finally realized bone regeneration. Conclusions In summary, our study proved that SR sealed channel could prevent the fibrous tissue from entering the fracture end and induce self-healing of long tubular bone through endochondral osteogenesis. In this process, the sealed environment provided by the SR channel was key, and this might indicate that the limit of self-healing of bone exceeded the previously thought. The translational potential of this article This study investigated a new concept to induce the self-healing of large bone defects. It could avoid trauma caused by autologous bone extraction and possible rejection reactions caused by bone graft materials. Further research based on this study, including the innovation of induction materials, might invent a new type of bone inducing production, which could bring convenience to patients. We believed that this study had significant meaning for the treatment of large bone defects in clinical practice.
Collapse
Affiliation(s)
- Feng Gu
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Ke Zhang
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Wan-an Zhu
- Department of Radiology, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhenjiang Sui
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Jiangbi Li
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaoping Xie
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Tiecheng Yu
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
7
|
Wu H, Tan J, Sun D, Wang X, Shen J, Wang S, Dai Q, Wei Z, Li G, Lin S, Luo F, Xie Z. Discovery of multipotent progenitor cells from human induced membrane: Equivalent to periosteum-derived stem cells in bone regeneration. J Orthop Translat 2023; 42:82-93. [PMID: 37705762 PMCID: PMC10495554 DOI: 10.1016/j.jot.2023.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/11/2023] [Accepted: 07/18/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND The periosteum stem cells (PSCs) plays a critical role in bone regeneration and defect reconstruction. Insertion of polymethyl methacrylate (PMMA) bone cement can form an induced membrane(IM) and showed promising strategy for bone defect reconstruction, the underlying mechanism remains unclear. Our study sought to determine whether IM-derived cells(IMDCs) versus PSCs have similar characteristics in bone regeneration. METHODS IM and periosteum were harvested from ten bone defect patients treated with PMMA, the IMDCs and PSCs were isolated respectively. Morphological, functional and molecular evaluation was performed and matched for comparison. RESULTS Both progenitor-like IMDCs and PSCs were successfully isolated. In vitro, we found IMDCs were similar to PSCs in morphology, colony forming capacity and expression of surface marker(CD90+, CD73+, CD105+, CD34-/CD45-). Meanwhile, these IMSCs displayed multipotency with chondrogenic, adipogenic and osteogenic differentiation, but differed in some IMSCs(3/10) population showing relatively poor osteogenic differentiation. The molecular profiles suggests that cell cycle and DNA replication signaling pathways were associated with these varying osteogenic potential. In vivo, we established a cell-based tissue-engineered bone by seeding IMDSs/PSCs to demineralized bone matrix (DBM) scaffold and demonstrated both IMDSs and PSCs enhanced bone regeneration in SCID mice bone defect model compared with DBM alone. CONCLUSION Our data demonstrated IM containing multipotent progenitor cells similar to that periosteum promoting bone regeneration, and indicated the existence of multiple subsets in osteogenic differentiation. Overall, the study provided a cellular and molecular insights in understanding the successful or failed outcome of bone defect healing.The translational potential of this article: This study confirmed IMDCs and PSCs share similar regeneration capacity and inform a translation potential of that cellular therapy applying IMDCs in bone defect repair.
Collapse
Affiliation(s)
- Hongri Wu
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
- Department of Orthopaedics, Navy 905 Hospital, Navy Medical University, Shanghai, PR China
| | - Jiulin Tan
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Dong Sun
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Xiaohua Wang
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Jie Shen
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Shulin Wang
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Qijie Dai
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Zhiyuan Wei
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Sien Lin
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Fei Luo
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Zhao Xie
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| |
Collapse
|
8
|
Frade BB, Dias RB, Gemini Piperni S, Bonfim DC. The role of macrophages in fracture healing: a narrative review of the recent updates and therapeutic perspectives. Stem Cell Investig 2023; 10:4. [PMID: 36817259 PMCID: PMC9936163 DOI: 10.21037/sci-2022-038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023]
Abstract
Objective This review addresses the latest advances in research on the role of macrophages in fracture healing, exploring their relationship with failures in bone consolidation and the perspectives for the development of advanced and innovative therapies to promote bone regeneration. Background The bone can fully restore its form and function after a fracture. However, the regenerative process of fracture healing is complex and is influenced by several factors, including macrophage activity. These cells have been found in the fracture site at all stages of bone regeneration, and their general depletion or the knockdown of receptors that mediate their differentiation, polarization, and/or function result in impaired fracture healing. Methods The literature search was carried out in the PubMed database, using combinations of the keywords "macrophage", "fracture healing, "bone regeneration", and "bone repair". Articles published within the last years (2017-2022) reporting evidence from in vivo long bone fracture healing experiments were included. Conclusions Studies published in the last five years on the role of macrophages in fracture healing strengthened the idea that what appears to be essential when it comes to a successful consolidation is the right balance between the M1/M2 populations, which have different but complementary roles in the process. These findings opened promising new avenues for the development of several macrophage-targeted therapies, including the administration of molecules and/or biomaterials intended to regulate macrophage differentiation and polarization, the local transplantation of macrophage precursors, and the use of exosomes to deliver signaling molecules that influence macrophage activities. However, more research is still warranted to better understand the diversity of macrophage phenotypes and their specific roles in each step of fracture healing and to decipher the key molecular mechanisms involved in the in vivo crosstalk between macrophages and other microenvironmental cell types, such as endothelial and skeletal stem/progenitor cells.
Collapse
Affiliation(s)
- Bianca Braga Frade
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil;,Postgraduation Program in Biological Sciences-Biophysics, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rhayra Braga Dias
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil;,Postgraduation Program in Morphological Sciences, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sara Gemini Piperni
- Laboratory of Biotechnology, Bioengineering and Nanostructured Biomaterials, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle Cabral Bonfim
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Manon J, Evrard R, Fievé L, Bouzin C, Magnin D, Xhema D, Darius T, Bonaccorsi-Riani E, Gianello P, Docquier PL, Schubert T, Lengelé B, Behets C, Cornu O. A New Osteogenic Membrane to Enhance Bone Healing: At the Crossroads between the Periosteum, the Induced Membrane, and the Diamond Concept. Bioengineering (Basel) 2023; 10:143. [PMID: 36829637 PMCID: PMC9952848 DOI: 10.3390/bioengineering10020143] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
The lack of viability of massive bone allografts for critical-size bone defect treatment remains a challenge in orthopedic surgery. The literature has reviewed the advantages of a multi-combined treatment with the synergy of an osteoconductive extracellular matrix (ECM), osteogenic stem cells, and growth factors (GFs). Questions are still open about the need for ECM components, the influence of the decellularization process on the latter, the related potential loss of function, and the necessity of using pre-differentiated cells. In order to fill in this gap, a bone allograft surrounded by an osteogenic membrane made of a decellularized collagen matrix from human fascia lata and seeded with periosteal mesenchymal stem cells (PMSCs) was analyzed in terms of de-/recellularization, osteogenic properties, PMSC self-differentiation, and angiogenic potential. While the decellularization processes altered the ECM content differently, the main GF content was decreased in soft tissues but relatively increased in hard bone tissues. The spontaneous osteogenic differentiation was necessarily obtained through contact with a mineralized bone matrix. Trying to deepen the knowledge on the complex matrix-cell interplay could further propel these tissue engineering concepts and lead us to provide the biological elements that allow bone integration in vivo.
Collapse
Affiliation(s)
- Julie Manon
- Morphology Lab (MORF), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 1200 Brussels, Belgium
- Transplantation and Experimental Surgery Lab (CHEX), IREC, UCLouvain, 1200 Brussels, Belgium
- Neuromusculoskeletal Lab (NMSK), IREC, UCLouvain, 1200 Brussels, Belgium
- Centre de Thérapie Cellulaire et Tissulaire Locomoteur, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Department of Orthopaedic and Trauma Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Robin Evrard
- Morphology Lab (MORF), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 1200 Brussels, Belgium
- Transplantation and Experimental Surgery Lab (CHEX), IREC, UCLouvain, 1200 Brussels, Belgium
- Neuromusculoskeletal Lab (NMSK), IREC, UCLouvain, 1200 Brussels, Belgium
- Centre de Thérapie Cellulaire et Tissulaire Locomoteur, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Department of Orthopaedic and Trauma Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Lies Fievé
- Morphology Lab (MORF), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 1200 Brussels, Belgium
| | - Caroline Bouzin
- Imaging Platform (2IP), IREC, UCLouvain, 1200 Brussels, Belgium
| | - Delphine Magnin
- Bio & Soft Matter (BSMA), Institute of Condensed Matter and Nanosciences (IMCN), UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | - Daela Xhema
- Transplantation and Experimental Surgery Lab (CHEX), IREC, UCLouvain, 1200 Brussels, Belgium
| | - Tom Darius
- Transplantation and Experimental Surgery Lab (CHEX), IREC, UCLouvain, 1200 Brussels, Belgium
- Department of Abdominal Surgery and Transplantation, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Eliano Bonaccorsi-Riani
- Transplantation and Experimental Surgery Lab (CHEX), IREC, UCLouvain, 1200 Brussels, Belgium
- Department of Abdominal Surgery and Transplantation, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Pierre Gianello
- Transplantation and Experimental Surgery Lab (CHEX), IREC, UCLouvain, 1200 Brussels, Belgium
| | - Pierre-Louis Docquier
- Neuromusculoskeletal Lab (NMSK), IREC, UCLouvain, 1200 Brussels, Belgium
- Department of Orthopaedic and Trauma Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Thomas Schubert
- Neuromusculoskeletal Lab (NMSK), IREC, UCLouvain, 1200 Brussels, Belgium
- Centre de Thérapie Cellulaire et Tissulaire Locomoteur, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Department of Orthopaedic and Trauma Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Benoît Lengelé
- Morphology Lab (MORF), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 1200 Brussels, Belgium
- Department of Plastic and Reconstructive Surgery, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium
| | - Catherine Behets
- Morphology Lab (MORF), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 1200 Brussels, Belgium
| | - Olivier Cornu
- Neuromusculoskeletal Lab (NMSK), IREC, UCLouvain, 1200 Brussels, Belgium
- Centre de Thérapie Cellulaire et Tissulaire Locomoteur, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Department of Orthopaedic and Trauma Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
10
|
Yufei T, Bingfeng W, Jiayi L, Hu L, Wenli L, Lin X. Distinct osteogenic effect of different periosteum derived cells via Hippo-YAP cascade signaling. Cell Cycle 2023; 22:183-199. [PMID: 35983614 PMCID: PMC9817120 DOI: 10.1080/15384101.2022.2111768] [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: 05/11/2022] [Revised: 07/27/2022] [Accepted: 08/06/2022] [Indexed: 01/11/2023] Open
Abstract
Periosteum is expected for bone repairing due to excellent regenerative potential. PDCs are the main source of cells for promoting bone repair. However, PDCs from different sites have been confirmed to be site specific due to their distinct embryonic origin and the methods of bone formation. Hippo-YAP pathway is proved to play a critical role in fate decision of mesenchymal stem cells. The effect of Hippo-YAP on PDCs has not been reported so far. Hence, we aim to explore the differences of PDCs from mandible and femur along with their possible responses to YAP signaling. mPDCs and fPDCs were obtained and tested through flow cytometry for identification. Follow-up results illustrated mPDCs was cubic shape and with better proliferation while fPDCs preferred slender cell shape with worse cell viability compared with mPDCs. mPDCs was superior to fPDCs in ALP activity, related mRNA expression and calcium deposits in late stage. Interestingly, downregulation of YAP promoted the ALP activity, related mRNA expression and calcium deposits of fPDCs while hindered that of mPDCs in vitro. Moreover, implant animal model in mandible and femur were constructed for evaluation in vivo. Histological results were similar to the results in vitro. We speculate this may result from their different embryonic origin and the way of bone formation. Taken together, results available suggested that mPDCs may serve as more optimal seed cells for tissue engineering compared with fPDCs; however, considering their different response to YAP signaling, to ensure sufficient YAP expression in mPDCs and appropriate declining YAP expression in fPDCs may establish better osteogenesis.
Collapse
Affiliation(s)
- Tang Yufei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wu Bingfeng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Jiayi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Long Hu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lai Wenli
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiang Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Wang D, Liu Y, Diao S, Shan L, Zhou J. Long Non-Coding RNAs Within Macrophage-Derived Exosomes Promote BMSC Osteogenesis in a Bone Fracture Rat Model. Int J Nanomedicine 2023; 18:1063-1083. [PMID: 36879890 PMCID: PMC9985426 DOI: 10.2147/ijn.s398446] [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: 11/25/2022] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
Purpose To investigate the effect of macrophage exosomal long non-coding (lnc)RNAs on bone mesenchymal stem cell (BMSC) osteogenesis and the associated mechanism. Methods Rat BMSCs and spleen macrophages were co-cultured with serum derived from the fracture microenvironment of rat tibia. BMSC osteogenesis was evaluated using Alizarin red staining and the expression of BMP-2, RUNX2, OPN, and OC mRNA. BMSC osteogenesis was evaluated after co-culture with macrophages stimulated using hypoxic conditions or colony-stimulating factor (CSF). The uptake of macrophage-derived exosomes by BMSCs was evaluated using the exosome uptake assay. High-throughput sequencing and bioinformatics analyses were performed to identify key lncRNAs in the macrophage exosomes. The effect of lncRNA expression levels on BMSC osteogenesis was also assessed using a lncRNA overexpression plasmid and siRNA technology. M1 and M2 macrophages were distinguished using flow cytometry and the key exosomal lncRNA was detected by in situ hybridization. Results In the fracture microenvironment, macrophages (stimulated using either hypoxia or CSF) significantly increased the osteogenic ability of BMSCs. We showed that BMSCs assimilated macrophage-derived vesicles and that the inhibition of exosomal secretion significantly attenuated the macrophage-mediated induction of BMSC osteogenesis. The hypoxia condition led to the up-regulation of 310 lncRNAs and the down-regulation of 575 lncRNAs in macrophage exosomes, while CSF stimulation caused the up-regulation of 557 lncRNAs and the down-regulation of 407 lncRNAs. In total, 108 lncRNAs were co-up-regulated and 326 lncRNAs were co-down-regulated under both conditions. We eventually identified LOC103691165 as a key lncRNA that promoted BMSC osteogenesis and was expressed at similar levels in both M1 and M2 macrophages. Conclusion In the fracture microenvironment, M1 and M2 macrophages promoted BMSC osteogenesis by secreting exosomes containing LOC103691165.
Collapse
Affiliation(s)
- Dong Wang
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Yang Liu
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Shuo Diao
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Lei Shan
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Junlin Zhou
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| |
Collapse
|
12
|
Hu Y, Huang J, Chen C, Wang Y, Hao Z, Chen T, Wang J, Li J. Strategies of Macrophages to Maintain Bone Homeostasis and Promote Bone Repair: A Narrative Review. J Funct Biomater 2022; 14:18. [PMID: 36662065 PMCID: PMC9864083 DOI: 10.3390/jfb14010018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Bone homeostasis (a healthy bone mass) is regulated by maintaining a delicate balance between bone resorption and bone formation. The regulation of physiological bone remodeling by a complex system that involves multiple cells in the skeleton is closely related to bone homeostasis. Loss of bone mass or repair of bone is always accompanied by changes in bone homeostasis. However, due to the complexity of bone homeostasis, we are currently unable to identify all the mechanisms that affect bone homeostasis. To date, bone macrophages have been considered a third cellular component in addition to osteogenic spectrum cells and osteoclasts. As confirmed by co-culture models or in vivo experiments, polarized or unpolarized macrophages interact with multiple components within the bone to ensure bone homeostasis. Different macrophage phenotypes are prone to resorption and formation of bone differently. This review comprehensively summarizes the mechanisms by which macrophages regulate bone homeostasis and concludes that macrophages can control bone homeostasis from osteoclasts, mesenchymal cells, osteoblasts, osteocytes, and the blood/vasculature system. The elaboration of these mechanisms in this narrative review facilitates the development of macrophage-based strategies for the treatment of bone metabolic diseases and bone defects.
Collapse
Affiliation(s)
- Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Jinghuan Huang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200000, China
| | - Chunying Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| |
Collapse
|
13
|
Tanios M, Brickman B, Cage E, Abbas K, Smith C, Atallah M, Baroi S, Lecka-Czernik B. Diabetes and Impaired Fracture Healing: A Narrative Review of Recent Literature. Curr Osteoporos Rep 2022; 20:229-239. [PMID: 35960475 DOI: 10.1007/s11914-022-00740-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF THE REVIEW Diabetes mellitus is a chronic metabolic disorder commonly encountered in orthopedic patients. Both type 1 and type 2 diabetes mellitus increase fracture risk and impair fracture healing. This review examines complex etiology of impaired fracture healing in diabetes. RECENT FINDINGS Recent findings point to several mechanisms leading to orthopedic complications in diabetes. Hyperglycemia and chronic inflammation lead to increased formation of advanced glycation end products and generation of reactive oxygen species, which in turn contribute to the disruption in osteoblast and osteoclast balance leading to decreased bone formation and heightening the risk of nonunion or delayed union as well as impaired fracture healing. The mechanisms attributing to this imbalance is secondary to an increase in pro-inflammatory mediators leading to premature resorption of callus cartilage and impaired bone formation due to compromised osteoblast differentiation and their apoptosis. Other mechanisms include disruption in the bone's microenvironment supporting different stages of healing process including hematoma and callus formation, and their resolution during bone remodeling phase. Complications of diabetes including peripheral neuropathy and peripheral vascular disease also contribute to the impairment of fracture healing. Certain diabetic drugs may have adverse effects on fracture healing. The pathophysiology of impaired fracture healing in diabetic patients is complex. This review provides an update of the most recent findings on how key mediators of bone healing are affected in diabetes.
Collapse
Affiliation(s)
- Mina Tanios
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| | - Bradley Brickman
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Emily Cage
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Kassem Abbas
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Cody Smith
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Marina Atallah
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sudipta Baroi
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Beata Lecka-Czernik
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
14
|
Tan WH, Winkler C. A non-disruptive and efficient knock-in approach allows fate tracing of resident osteoblast progenitors during repair of vertebral lesions in medaka. Development 2022; 149:275483. [DOI: 10.1242/dev.200238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 05/11/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
During bone development and repair, osteoblasts are recruited to bone deposition sites. To identify the origin of recruited osteoblasts, cell lineage tracing using Cre/loxP recombination is commonly used. However, a confounding factor is the use of transgenic Cre drivers that do not accurately recapitulate endogenous gene expression or the use of knock-in Cre drivers that alter endogenous protein activity or levels. Here, we describe a CRISPR/Cas9 homology-directed repair knock-in approach that allows efficient generation of Cre drivers controlled by the endogenous gene promoter. In addition, a self-cleaving peptide preserves the reading frame of the endogenous protein. Using this approach, we generated col10a1p2a-CreERT2 knock-in medaka and show that tamoxifen-inducible CreERT2 efficiently recombined loxP sites in col10a1 cells. Similar knock-in efficiencies were obtained when two unrelated loci (osr1 and col2a1a) were targeted. Using live imaging, we traced the fate of col10a1 osteoblast progenitors during bone lesion repair in the medaka vertebral column. We show that col10a1 cells at neural arches represent a mobilizable cellular source for bone repair. Together, our study describes a previously unreported strategy for precise cell lineage tracing via efficient and non-disruptive knock-in of Cre.
Collapse
Affiliation(s)
- Wen Hui Tan
- National University of Singapore Department of Biological Sciences and Centre for Bioimaging Sciences , , Singapore 117543 , Singapore
| | - Christoph Winkler
- National University of Singapore Department of Biological Sciences and Centre for Bioimaging Sciences , , Singapore 117543 , Singapore
| |
Collapse
|
15
|
One of the Primary Functions of Tissue-Resident Pluripotent Pericytes Cells May Be to Regulate Normal Organ Growth and Maturation: Implications for Attempts to Repair Tissues Later in Life. Int J Mol Sci 2022; 23:ijms23105496. [PMID: 35628309 PMCID: PMC9146368 DOI: 10.3390/ijms23105496] [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: 04/28/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/04/2022] Open
Abstract
Adult mesenchymal stem cells were reported more than 30 years ago. Since then, their potential to repair and regenerate damaged or diseased tissues has been studied intensively in both preclinical models and human trials. Most of the need for such tissue repair/regeneration is in older populations, so much of the effort has been performed with autologous cells in older patients. However, success has been difficult to achieve. In the literature, it has been noted that such progenitor cells from younger individuals often behave with more vigorous activity and are functionally enhanced compared to those from older individuals or animals. In addition, cells with the characteristics of mesenchymal stem cells or pluripotent mesenchymal regulatory cells exist in nearly all tissues and organs as pericytes since fetal life. Such evidence raises the possibility that one of the primary roles of these organ-specific cells is to regulate organ growth and maturation, and then subsequently play a role in the maintenance of organ integrity. This review will discuss the evidence to support this concept and the implications of such a concept regarding the use of these progenitor cells for the repair and regeneration of tissues damaged by injury or disease later in life. For the latter, it may be necessary to return the organ-specific progenitor cells to the functional state that contributed to their effectiveness during growth and maturation rather than attempting to use them after alterations imposed during the aging process have been established and their function compromised.
Collapse
|
16
|
Lobov A, Malashicheva A. Osteogenic differentiation: a universal cell program of heterogeneous mesenchymal cells or a similar extracellular matrix mineralizing phenotype? BIOLOGICAL COMMUNICATIONS 2022; 67. [DOI: 10.21638/spbu03.2022.104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Despite the popularity of mesenchymal stem cells (MSCs), many fundamental aspects of their physiology still have not been understood. The information accumulated to date argues that MSCs from different sources vary in their differentiation potential and, probably, in molecular mechanisms of trilineage differentiation. Therefore, this review consists of two parts. Firstly, we focus on the data on inter- and intra-source variation of MSCs. We discuss in detail MSC variation at the single-cell level and direct omics comparison of MSCs from four main tissue sources: bone marrow, adipose tissue, umbilical cord and tooth. MSCs from all tissues represent heterogeneous populations in vivo with sub-populational structures reflecting their functional role in the tissue. After in vitro cultivation MSCs lose their natural heterogeneity, but obtain a new one, which might be regarded as a cultivation artifact. Nevertheless, MSCs from various sources still keep their functional differences after in vitro cultivation. In the second part of the review, we discuss how these differences influence molecular mechanisms of osteogenic differentiation. We highlight at least one subtype of mesenchymal cells differentiation with matrix mineralization — odontoblastic differentiation. We also discuss differences in molecular mechanisms of pathological heterotopic osteogenic differentiation of valve interstitial and tumor cells, but these assumptions need additional empirical confirmation. Finally, we observe differences in osteogenic differentiation molecular mechanisms of several MSC types and argue that this differentiation might be influenced by the cell context. Nevertheless, bone marrow and adipose MSCs seem to undergo osteogenic differentiation similarly, by the same mechanisms.
Collapse
|
17
|
Bragdon BC, Bennie A, Molinelli A, Liu Y, Gerstenfeld LC. Post natal expression of Prx1 labels appendicular restricted progenitor cell populations of multiple tissues. J Cell Physiol 2022; 237:2550-2560. [PMID: 35338481 PMCID: PMC9133217 DOI: 10.1002/jcp.30728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 12/26/2022]
Abstract
Currently, there is no consensus whether there is a single or multiple postnatal stem cell population(s) that contribute to skeletal homeostasis and postnatal bone formation. A known population of cells that express Prx1 contributes to postnatal bone formation. Prx1 expression also connotes calvaria and appendicular tissues during embryonic development. A transgenic tamoxifen inducible Prx1 reporter mouse was used for lineage tracking, to characterize the postnatal contribution of Prx1 expressing cells in skeletal homeostasis and bone formation. Under homeostatic conditions Prx1 labeling gave rise to a transient yet rapid turnover cell population at the periosteal and endosteal surfaces, along muscle fibers, and within the medial layers of vessels both within the muscle and marrow compartments of the appendicular skeleton. Fracture and ectopic bone formation of both fore and hind limbs showed recruitment and expansion of Prx1-derived cells in newly formed bone tissues. Prx1 labeled cells were limited or absent at axial skeletal sites during both homeostasis and after induction of bone formation. Last, Prx1-derived cells differentiated into multiple cell lineages including vascular smooth muscle, adipose, cartilage, and bone cells. These results show that Prx1 expression retained its embryonic tissue specification and connotes a stem/progenitor cell populations of mesenchymal tissue progenitors.
Collapse
Affiliation(s)
- Beth C Bragdon
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Andrew Bennie
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Amanda Molinelli
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yu Liu
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
18
|
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
|
19
|
Wang L, Yao L, Duan H, Yang F, Lin M, Zhang R, He Z, Ahn J, Fan Y, Qin L, Gong Y. Plasminogen Regulates Fracture Repair by Promoting the Functions of Periosteal Mesenchymal Progenitors. J Bone Miner Res 2021; 36:2229-2242. [PMID: 34378815 PMCID: PMC8865375 DOI: 10.1002/jbmr.4423] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/06/2022]
Abstract
Defective or insufficient bone repair and regeneration are common in patients as a result of major trauma or severe disease. Cell therapy with periosteal mesenchymal progenitors, which can be limited in severe injury, serves as a promising approach; however, its efficacy is limited due to a repair-hostile ischemic tissue microenvironment after traumatic fracture. Here we report that plasminogen (Plg), a factor that is upregulated in these environments, is critical for fracture healing. Plg knockout mice had impaired trabecular and cortical bone structure and exhibited delayed and incomplete fracture healing. Interestingly, Plg deficiency greatly reduced the thickness of expanded periosteum, suggesting a role of Plg in periosteal mesenchymal progenitor-mediated bone repair. In culture, Plg increased cell proliferation and migration in periosteal mesenchymal progenitors and inhibited cell death under ischemic conditions. Mechanistically, we revealed that Plg cleaved and activated Cyr61 to regulate periosteal progenitor function. Thus, our study uncovers a cellular mechanism underlying fracture healing, by which Plg activates Cyr61 to promote periosteal progenitor proliferation, survival, and migration and improves bone repair after fracture. Targeting Plg may offer a rational and effective therapeutic opportunity for improving fracture healing. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Luqiang Wang
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Orthopaedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lutian Yao
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, China
| | - Hao Duan
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Neurosurgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fan Yang
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maohuan Lin
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rongxin Zhang
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenqiang He
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaimo Ahn
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Fan
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanqing Gong
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
20
|
Kato Y, Yoshida S, Kato T. New insights into the role and origin of pituitary S100β-positive cells. Cell Tissue Res 2021; 386:227-237. [PMID: 34550453 DOI: 10.1007/s00441-021-03523-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/07/2021] [Indexed: 01/16/2023]
Abstract
In the anterior pituitary, S100β protein (S100β) has been assumed to be a marker of folliculo-stellate cells, which are one of the non-hormone-producing cells existing in the parenchyma of the adult anterior lobe and are composed of subpopulations with various functions. However, recent accumulating studies on S100β-positive cells, including non-folliculo-stellate cells lining the marginal cell layer (MCL), have shown the novel aspect that most S100β-positive cells in the MCL and parenchyma of the adult anterior lobe are positive for sex determining region Y-box 2 (SOX2), a marker of pituitary stem/progenitor cells. From the viewpoint of SOX2-positive cells, the majority of these cells in the MCL and in the parenchyma are positive for S100β, suggesting that S100β plays a role in the large population of stem/progenitor cells in the anterior lobe of the adult pituitary. Reportedly, S100β/SOX2-double positive cells are able to differentiate into hormone-producing cells and various types of non-hormone-producing cells. Intriguingly, it has been demonstrated that extra-pituitary lineage cells invade the pituitary gland during prenatal pituitary organogenesis. Among them, two S100β-positive populations have been identified: one is SOX2-positive population which invades at the late embryonic period through the pituitary stalk and another is a SOX2-negative population that invades at the middle embryonic period through Atwell's recess. These two populations are likely the substantive origin of S100β-positive cells in the postnatal anterior pituitary, while S100β-positive cells emerging from oral ectoderm-derived cells remain unclear.
Collapse
Affiliation(s)
- Yukio Kato
- Institute for Endocrinology, Meiji University, 1-1-1 Higashi-mita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan.
| | - Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takako Kato
- Institute for Endocrinology, Meiji University, 1-1-1 Higashi-mita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| |
Collapse
|
21
|
Wildemann B, Ignatius A, Leung F, Taitsman LA, Smith RM, Pesántez R, Stoddart MJ, Richards RG, Jupiter JB. Non-union bone fractures. Nat Rev Dis Primers 2021; 7:57. [PMID: 34354083 DOI: 10.1038/s41572-021-00289-8] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 11/09/2022]
Abstract
The human skeleton has remarkable regenerative properties, being one of the few structures in the body that can heal by recreating its normal cellular composition, orientation and mechanical strength. When the healing process of a fractured bone fails owing to inadequate immobilization, failed surgical intervention, insufficient biological response or infection, the outcome after a prolonged period of no healing is defined as non-union. Non-union represents a chronic medical condition not only affecting function but also potentially impacting the individual's psychosocial and economic well-being. This Primer provides the reader with an in-depth understanding of our contemporary knowledge regarding the important features to be considered when faced with non-union. The normal mechanisms involved in bone healing and the factors that disrupt the normal signalling mechanisms are addressed. Epidemiological considerations and advances in the diagnosis and surgical therapy of non-union are highlighted and the need for greater efforts in basic, translational and clinical research are identified.
Collapse
Affiliation(s)
- Britt Wildemann
- Experimental Trauma Surgery, Department of Trauma, Hand and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany. .,Julius Wolff Institute and BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University, Ulm, Baden Württemberg, Germany
| | - Frankie Leung
- Department of Orthopaedics and Traumatology, Queen Mary Hospital, the University of Hong Kong, Hong Kong, Hong Kong
| | - Lisa A Taitsman
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | - R Malcolm Smith
- Orthopedic trauma service, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rodrigo Pesántez
- Departamento de Ortopedia Y Traumatología Fundación Santa Fé de Bogotá - Universidad de los Andes, Bogotá, Colombia
| | | | | | - Jesse B Jupiter
- Department of Orthopaedic surgery, Massachussets General Hospital, Boston, MA, USA.
| |
Collapse
|
22
|
Abstract
This chapter describes the methods of isolation of mouse periosteal progenitor cells. There are three basic methods utilized. The bone grafting method was developed utilizing the fracture healing process to expand the progenitor populations. Bone capping methods requires enzymatic digestion and purification of cells from the native periosteum, while the Egression/Explant method requires the least manipulation with placement of cortical bone fragments with attached periosteum in a culture dish. Various cell surface antibodies have been employed over the years to characterize periosteum derived progenitor cells, but the most consistent minimal criteria was recommended by the International Society for Cellular Therapy. Confirmation of the multipotent status of these isolated cells can be achieved by differentiation into the three basic mesodermal lineages in vitro.
Collapse
|
23
|
Shen L, Xiao Y, Xie H, Zhao H, Luo T, Liu L, Pan X. A naturally derived small molecule NDSM253 inhibits IKK1 to suppress inflammation response and promote bone healing after fracture. Am J Transl Res 2021; 13:24-37. [PMID: 33527006 PMCID: PMC7847520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/04/2020] [Indexed: 06/12/2023]
Abstract
Bone fracture induces an acute inflammatory response in the resident and peripheral monocyte/macrophage cells. Excessive amounts of proinflammatory cytokines can cause severe tissue damage and inhibit bone healing. The proinflammatory cytokine genes are mainly controlled by TLR4/NF-κB (Toll-like receptor 4/Nuclear factor κB). Thus, targeting the molecules in this signaling pathway to decrease the expression of proinflammatory cytokines is an effective strategy to inhibit the inflammatory response. Herein, we identified a naturally derived small molecule NDSM253 that specifically inhibited IKKα (Inhibitor of NF-κB kinase subunit-alpha), a critical component of TLR4/NF-κB signaling. Biochemically, NDMS253 decreased phosphorylation of IκB (Inhibitor of NF-κB), thereby increasing the binding of IκB-NF-κB and suppressing the proinflammatory cytokine gene expression. NDMS253 showed a much stronger inhibitory effect on proinflammatory cytokine gene expression than did the known IKK inhibitors, including ACHP (2-Amino-6-[2-(cyclopropylmethoxy)-6-hydroxyphenyl]-4-(4-piperidinyl)-3-pyridinecarbonitrile), IKK16, and Amlexanox. Administration of these IKK inhibitors in a mouse femoral fracture model showed that NDSM253 suppressed proinflammatory cytokine genes, thereby promoting bone healing, while the other three IKK inhibitors showed a weaker improvement of both bone healing and circulating proinflammatory cytokines. Collectively, our data suggested that NDSM253 might be an effective inhibitor of IKKα that could inhibit inflammatory cytokine action in bone injury.
Collapse
Affiliation(s)
- Liqi Shen
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Yun Xiao
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Hui Xie
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Hongbin Zhao
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Tao Luo
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Lin Liu
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Xuekun Pan
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| |
Collapse
|
24
|
Dingle M, Fernicola SD, de Vasconcellos JF, Zicari S, Daniels C, Dunn JC, Dimtchev A, Nesti LJ. Characterization of traumatized muscle-derived multipotent progenitor cells from low-energy trauma. Stem Cell Res Ther 2021; 12:6. [PMID: 33407850 PMCID: PMC7788846 DOI: 10.1186/s13287-020-02038-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022] Open
Abstract
Background Multipotent progenitor cells have been harvested from different human tissues, including the bone marrow, adipose tissue, and umbilical cord blood. Previously, we identified a population of mesenchymal progenitor cells (MPCs) isolated from the traumatized muscle of patients undergoing reconstructive surgery following a war-related blast injury. These cells demonstrated the ability to differentiate into multiple mesenchymal lineages. While distal radius fractures from a civilian setting have a much lower injury mechanism (low-energy trauma), we hypothesized that debrided traumatized muscle near the fracture site would contain multipotent progenitor cells with the ability to differentiate and regenerate the injured tissue. Methods The traumatized muscle was debrided from the pronator quadratus in patients undergoing open reduction and internal fixation for a distal radius fracture at the Walter Reed National Military Medical Center. Using a previously described protocol for the isolation of MPCs from war-related extremity injuries, cells were harvested from the low-energy traumatized muscle samples and expanded in culture. Isolated cells were characterized by flow cytometry and q-RT-PCRs and induced to adipogenic, osteogenic, and chondrogenic differentiation. Downstream analyses consisted of lineage-specific staining and q-RT-PCR. Results Cells isolated from low-energy traumatized muscle samples were CD73+, CD90+, and CD105+ that are the characteristic of adult human mesenchymal stem cells. These cells expressed high levels of the stem cell markers OCT4 and NANOG 1-day after isolation, which was dramatically reduced over-time in monolayer culture. Following induction, lineage-specific markers were demonstrated by each specific staining and confirmed by gene expression analysis, demonstrating the ability of these cells to differentiate into adipogenic, osteogenic, and chondrogenic lineages. Conclusions Adult multipotent progenitor cells are an essential component for the success of regenerative medicine efforts. While MPCs have been isolated and characterized from severely traumatized muscle from high-energy injuries, here, we report that cells with similar characteristics and multipotential capacity have been isolated from the tissue that was exposed to low-energy, community trauma.
Collapse
Affiliation(s)
- Marvin Dingle
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Department of Orthopaedic Surgery, Walter Reed National Military Medical Center, 4801 Rockville Pike, Bethesda, MD, 20889, USA
| | - Stephen D Fernicola
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Department of Orthopaedic Surgery, Walter Reed National Military Medical Center, 4801 Rockville Pike, Bethesda, MD, 20889, USA
| | - Jaira F de Vasconcellos
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD, 20817, USA
| | - Sonia Zicari
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD, 20817, USA
| | - Christopher Daniels
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Department of Orthopaedic Surgery, Walter Reed National Military Medical Center, 4801 Rockville Pike, Bethesda, MD, 20889, USA
| | - John C Dunn
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,William Beaumont Army Medical Center, 5005 N Piedras St, El Paso, TX, 79920, USA
| | - Alexander Dimtchev
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD, 20817, USA
| | - Leon J Nesti
- Clinical and Experimental Orthopaedics, Department of Surgery, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,Department of Orthopaedic Surgery, Walter Reed National Military Medical Center, 4801 Rockville Pike, Bethesda, MD, 20889, USA.
| |
Collapse
|
25
|
Kegelman CD, Nijsure MP, Moharrer Y, Pearson HB, Dawahare JH, Jordan KM, Qin L, Boerckel JD. YAP and TAZ Promote Periosteal Osteoblast Precursor Expansion and Differentiation for Fracture Repair. J Bone Miner Res 2021; 36:143-157. [PMID: 32835424 PMCID: PMC7988482 DOI: 10.1002/jbmr.4166] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
In response to bone fracture, periosteal progenitor cells proliferate, expand, and differentiate to form cartilage and bone in the fracture callus. These cellular functions require the coordinated activation of multiple transcriptional programs, and the transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) regulate osteochondroprogenitor activation during endochondral bone development. However, recent observations raise important distinctions between the signaling mechanisms used to control bone morphogenesis and repair. Here, we tested the hypothesis that YAP and TAZ regulate osteochondroprogenitor activation during endochondral bone fracture healing in mice. Constitutive YAP and/or TAZ deletion from Osterix-expressing cells impaired both cartilage callus formation and subsequent mineralization. However, this could be explained either by direct defects in osteochondroprogenitor differentiation after fracture or by developmental deficiencies in the progenitor cell pool before fracture. Consistent with the second possibility, we found that developmental YAP/TAZ deletion produced long bones with impaired periosteal thickness and cellularity. Therefore, to remove the contributions of developmental history, we next generated adult onset-inducible knockout mice (using Osx-CretetOff ) in which YAP and TAZ were deleted before fracture but after normal development. Adult onset-induced YAP/TAZ deletion had no effect on cartilaginous callus formation but impaired bone formation at 14 days post-fracture (dpf). Earlier, at 4 dpf, adult onset-induced YAP/TAZ deletion impaired the proliferation and expansion of osteoblast precursor cells located in the shoulder of the callus. Further, activated periosteal cells isolated from this region at 4 dpf exhibited impaired osteogenic differentiation in vitro upon YAP/TAZ deletion. Finally, confirming the effects on osteoblast function in vivo, adult onset-induced YAP/TAZ deletion impaired bone formation in the callus shoulder at 7 dpf before the initiation of endochondral ossification. Together, these data show that YAP and TAZ promote the expansion and differentiation of periosteal osteoblast precursors to accelerate bone fracture healing. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Christopher D Kegelman
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasaman Moharrer
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Hope B Pearson
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - James H Dawahare
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Kelsey M Jordan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
26
|
Jin Y, Sun X, Pei F, Zhao Z, Mao J. Wnt16 signaling promotes osteoblast differentiation of periosteal derived cells in vitro and in vivo. PeerJ 2020; 8:e10374. [PMID: 33282557 PMCID: PMC7694570 DOI: 10.7717/peerj.10374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Background Periosteum plays critical roles in de novo bone formation and fracture repair. Wnt16 has been regarded as a key regulator in periosteum bone formation. However, the role of Wnt16 in periosteum derived cells (PDCs) osteogenic differentiation remains unclear. The study goal is to uncover whether and how Wnt16 acts on the osteogenesis of PDCs. Methods We detected the variation of Wnt16 mRNA expression in PDCs, which were isolated from mouse femur and identified by flow cytometry, cultured in osteogenic medium for 14 days, then knocked down and over-expressed Wnt16 in PDCs to analysis its effects in osteogenesis. Further, we seeded PDCs (Wnt16 over-expressed/vector) in β-tricalcium phosphate cubes, and transplanted this complex into a critical size calvarial defect. Lastly, we used immunofluorescence, Topflash and NFAT luciferase reporter assay to study the possible downstream signaling pathway of Wnt16. Results Wnt16 mRNA expression showed an increasing trend in PDCs under osteogenic induction for 14 days. Wnt16 shRNA reduced mRNA expression of Runx2, collage type I (Col-1) and osteocalcin (OCN) after 7 days of osteogenic induction, as well as alizarin red staining intensity after 21days. Wnt16 also increased the mRNA expression of Runx2 and OCN and the protein production of Runx2 and Col-1 after 2 days of osteogenic stimulation. In the orthotopic transplantation assay, more bone volume, trabecula number and less trabecula space were found in Wnt16 over-expressed group. Besides, in the newly formed tissue Brdu positive area was smaller and Col-1 was larger in Wnt16 over-expressed group compared to the control group. Finally, Wnt16 upregulated CTNNB1/β-catenin expression and its nuclear translocation in PDCs, also increased Topflash reporter luciferase activity. By contrast, Wnt16 failed to increase NFAT reporter luciferase activity. Conclusion Together, Wnt16 plays a positive role in regulating PDCs osteogenesis, and Wnt16 may have a potential use in improving bone regeneration.
Collapse
Affiliation(s)
- Ying Jin
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyan Sun
- Stomatological Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Pei
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jeremy Mao
- Columbia University, Center for Craniofacial Regeneration, New York, NY, United States of America
| |
Collapse
|
27
|
Cao Y, Buckels EJ, Matthews BG. Markers for Identification of Postnatal Skeletal Stem Cells In Vivo. Curr Osteoporos Rep 2020; 18:655-665. [PMID: 33034805 DOI: 10.1007/s11914-020-00622-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The adult skeleton contains stem cells involved in growth, homeostasis, and healing. Mesenchymal or skeletal stem cells are proposed to provide precursors to osteoblasts, chondrocytes, marrow adipocytes, and stromal cells. We review the evidence for existence and functionality of different skeletal stem cell pools, and the tools available for identifying or targeting these populations in mouse and human tissues. RECENT FINDINGS Lineage tracing and single cell-based techniques in mouse models indicate that multiple pools of stem cells exist in postnatal bone. These include growth plate stem cells, stem and progenitor cells in the diaphysis, reticular cells that only form bone in response to injury, and injury-responsive periosteal stem cells. New staining protocols have also been described for prospective isolation of human skeletal stem cells. Several populations of postnatal skeletal stem and progenitor cells have been identified in mice, and we have an increasing array of tools to target these cells. Most Cre models lack a high degree of specificity to define single populations. Human studies are less advanced and require further efforts to refine methods for identifying stem and progenitor cells in adult bone.
Collapse
Affiliation(s)
- Ye Cao
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Emma J Buckels
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Brya G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand.
| |
Collapse
|
28
|
Xia C, Ge Q, Fang L, Yu H, Zou Z, Zhang P, Lv S, Tong P, Xiao L, Chen D, Wang PE, Jin H. TGF-β/Smad2 signalling regulates enchondral bone formation of Gli1 + periosteal cells during fracture healing. Cell Prolif 2020; 53:e12904. [PMID: 32997394 PMCID: PMC7653269 DOI: 10.1111/cpr.12904] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/20/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Most bone fracture heals through enchondral bone formation that relies on the involvement of periosteal progenitor cells. However, the identity of periosteal progenitor cells and the regulatory mechanism of their proliferation and differentiation remain unclear. The aim of this study was to investigate whether Gli1-CreERT2 can identify a population of murine periosteal progenitor cells and the role of TGF-β signalling in periosteal progenitor cells on fracture healing. MATERIALS AND METHODS Double heterozygous Gli1-CreERT2 ;Rosa26-tdTomatoflox/wt mice were sacrificed at different time points for tracing the fate of Gli1+ cells in both intact and fracture bone. Gli1-CreERT2 -mediated Tgfbr2 knockout (Gli1-CreERT2 ;Tgfbr2flox/flox ) mice were subjected to fracture surgery. At 4, 7, 10, 14 and 21 days post-surgery, tibia samples were harvested for tissue analyses including μCT, histology, real-time PCR and immunofluorescence staining. RESULTS Through cell lineage-tracing experiments, we have revealed that Gli1-CreER T2 can be used to identify a subpopulation of periosteal progenitor cells in vivo that persistently reside in periosteum and contribute to osteochondral elements during fracture repair. During the healing process, TGF-β signalling is continually activated in the reparative Gli1+ periosteal cells. Conditional knockout of Tgfbr2 in these cells leads to a delayed and impaired enchondral bone formation, at least partially due to the reduced proliferation and chondrogenic and osteogenic differentiation of Gli1+ periosteal cells. CONCLUSIONS TGF-β signalling plays an essential role on fracture repair via regulating enchondral bone formation process of Gli1+ periosteal cells.
Collapse
Affiliation(s)
- Chenjie Xia
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopedic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Qinwen Ge
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liang Fang
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huan Yu
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhen Zou
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peng Zhang
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuaijie Lv
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Luwei Xiao
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ping-Er Wang
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongting Jin
- Institute of Orthopadics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
29
|
Ortinau LC, Wang H, Lei K, Deveza L, Jeong Y, Hara Y, Grafe I, Rosenfeld SB, Lee D, Lee B, Scadden DT, Park D. Identification of Functionally Distinct Mx1+αSMA+ Periosteal Skeletal Stem Cells. Cell Stem Cell 2020; 25:784-796.e5. [PMID: 31809737 DOI: 10.1016/j.stem.2019.11.003] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/11/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022]
Abstract
The periosteum is critical for bone maintenance and healing. However, the in vivo identity and specific regulatory mechanisms of adult periosteum-resident skeletal stem cells are unknown. Here, we report animal models that selectively and durably label postnatal Mx1+αSMA+ periosteal stem cells (P-SSCs) and establish that P-SSCs are a long-term repopulating, functionally distinct SSC subset responsible for lifelong generation of periosteal osteoblasts. P-SSCs rapidly migrate toward an injury site, supply osteoblasts and chondrocytes, and recover new periosteum. Notably, P-SSCs specifically express CCL5 receptors, CCR3 and CCR5. Real-time intravital imaging revealed that the treatment with CCL5 induces P-SSC migration in vivo and bone healing, while CCL5/CCR5 deletion, CCR5 inhibition, or local P-SSC ablation reduces osteoblast number and delays bone healing. Human periosteal cells express CCR5 and undergo CCL5-mediated migration. Thus, the adult periosteum maintains genetically distinct SSC subsets with a CCL5-dependent migratory mechanism required for bone maintenance and injury repair.
Collapse
Affiliation(s)
- Laura C Ortinau
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hamilton Wang
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Kevin Lei
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Lorenzo Deveza
- Department of Orthopedic Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Youngjae Jeong
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yannis Hara
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ingo Grafe
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Scott B Rosenfeld
- Texas Children's Hospital, 6701 Fannin Street, Houston, TX 77030, USA
| | - Dongjun Lee
- Department of Convergence of Medical Science, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Brendan Lee
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - David T Scadden
- Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Dongsu Park
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Desai S, Jayasuriya CT. Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair. Bioengineering (Basel) 2020; 7:E86. [PMID: 32759659 PMCID: PMC7552784 DOI: 10.3390/bioengineering7030086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Harnessing adult mesenchymal stem/progenitor cells to stimulate skeletal tissue repair is a strategy that is being actively investigated. While scientists continue to develop creative and thoughtful ways to utilize these cells for tissue repair, the vast majority of these methodologies can ultimately be categorized into two main approaches: (1) Facilitating the recruitment of endogenous host cells to the injury site; and (2) physically administering into the injury site cells themselves, exogenously, either by autologous or allogeneic implantation. The aim of this paper is to comprehensively review recent key literature on the use of these two approaches in stimulating healing and repair of different skeletal tissues. As expected, each of the two strategies have their own advantages and limitations (which we describe), especially when considering the diverse microenvironments of different skeletal tissues like bone, tendon/ligament, and cartilage/fibrocartilage. This paper also discusses stem/progenitor cells commonly used for repairing different skeletal tissues, and it lists ongoing clinical trials that have risen from the implementation of these cells and strategies. Lastly, we discuss our own thoughts on where the field is headed in the near future.
Collapse
Affiliation(s)
| | - Chathuraka T. Jayasuriya
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and the Rhode Island Hospital, Providence, RI 02903, USA;
| |
Collapse
|
31
|
Hellwinkel JE, Miclau T, Provencher MT, Bahney CS, Working ZM. The Life of a Fracture: Biologic Progression, Healing Gone Awry, and Evaluation of Union. JBJS Rev 2020; 8:e1900221. [PMID: 32796195 PMCID: PMC11147169 DOI: 10.2106/jbjs.rvw.19.00221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
New knowledge about the molecular biology of fracture-healing provides opportunities for intervention and reduction of risk for specific phases that are affected by disease and medications. Modifiable and nonmodifiable risk factors can prolong healing, and the informed clinician should optimize each patient to provide the best chance for union. Techniques to monitor progression of fracture-healing have not changed substantially over time; new objective modalities are needed.
Collapse
Affiliation(s)
- Justin E Hellwinkel
- Department of Orthopedic Surgery, New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Theodore Miclau
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Matthew T Provencher
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Chelsea S Bahney
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Zachary M Working
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
- Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
32
|
Carluccio M, Ziberi S, Zuccarini M, Giuliani P, Caciagli F, Di Iorio P, Ciccarelli R. Adult mesenchymal stem cells: is there a role for purine receptors in their osteogenic differentiation? Purinergic Signal 2020; 16:263-287. [PMID: 32500422 DOI: 10.1007/s11302-020-09703-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
The role played by mesenchymal stem cells (MSCs) in contributing to adult tissue homeostasis and damage repair thanks to their differentiation capabilities has raised a great interest, mainly in bone regenerative medicine. The growth/function of these undifferentiated cells of mesodermal origin, located in specialized structures (niches) of differentiated organs is influenced by substances present in this microenvironment. Among them, ancestral and ubiquitous molecules such as adenine-based purines, i.e., ATP and adenosine, may be included. Notably, extracellular purine concentrations greatly increase during tissue injury; thus, MSCs are exposed to effects mediated by these agents interacting with their own receptors when they act/migrate in vivo or are transplanted into a damaged tissue. Here, we reported that ATP modulates MSC osteogenic differentiation via different P2Y and P2X receptors, but data are often inconclusive/contradictory so that the ATP receptor importance for MSC physiology/differentiation into osteoblasts is yet undetermined. An exception is represented by P2X7 receptors, whose expression was shown at various differentiation stages of bone cells resulting essential for differentiation/survival of both osteoclasts and osteoblasts. As well, adenosine, usually derived from extracellular ATP metabolism, can promote osteogenesis, likely via A2B receptors, even though findings from human MSCs should be implemented and confirmed in preclinical models. Therefore, although many data have revealed possible effects caused by extracellular purines in bone healing/remodeling, further studies, hopefully performed in in vivo models, are necessary to identify defined roles for these compounds in favoring/increasing the pro-osteogenic properties of MSCs and thereby their usefulness in bone regenerative medicine.
Collapse
Affiliation(s)
- Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy.,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy
| | - Sihana Ziberi
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy.,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Francesco Caciagli
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy. .,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy. .,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy.
| |
Collapse
|
33
|
Hu S, Ge Q, Xia C, Ying J, Ruan H, Shi Z, Xu R, Xu T, Lv S, Fang L, Zou Z, Xu H, Xiao L, Tong P, Wang PE, Jin H. Bushenhuoxue formula accelerates fracture healing via upregulation of TGF-β/Smad2 signaling in mesenchymal progenitor cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153256. [PMID: 32534359 DOI: 10.1016/j.phymed.2020.153256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/10/2020] [Accepted: 05/29/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Although Bushenhuoxue formula (BSHXF) is successfully used as a non-traumatic therapy in treating bone fracture in China, the molecular mechanism underlying its effects remains poorly understood. PURPOSE The present study aims to explore the therapeutic effects of BSHXF on fracture healing in mice and the underlying mechanism. METHODS We performed unilateral open transverse tibial fracture procedure in C57BL/6 mice which were treated with or without BSHXF. Fracture callus tissues were collected and analyzed by X-ray, micro-CT, biomechanical testing, histopathology and quantitative gene expression analysis. Tibial fracture procedure was also performed in Cre-negative and Gli1-CreER; Tgfbr2flox/flox conditional knockout (KO) mice (Tgfbr2Gli1ER) to determine if BSHXF enhances fracture healing in a TGF-β-dependent manner. In addition, scratch-wound assay and cell counting kit-8 (CCK-8) assay were used to evaluate the effect of BSHXF on cell migration and cell proliferation in C3H10T1/2 mesenchymal stem cells, respectively. RESULTS BSHXF promoted endochondral ossification and enhanced bone strength in wild-type (WT) or Cre- control mice. In contrast, BSHXF failed to promote bone fracture healing in Tgfbr2Gli1ER conditional KO mice. In the mice receiving BSHXF treatment, TGF-β/Smad2 signaling was significantly activated. Moreover, BSHXF enhanced cell migration and cell proliferation in C3H10T1/2 cells, which was strongly attenuated by the small molecule inhibitor SB525334 against TGF-β type I receptor. CONCLUSION These data demonstrated that BSHXF promotes fracture healing by activating TGF-β/Smad2 signaling. BSHXF may be used as a type of alternative medicine for the treatment of bone fracture healing.
Collapse
Affiliation(s)
- Songfeng Hu
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; Department of Orthopaedics and Traumatology, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing 312000, Zhejiang, China
| | - Qinwen Ge
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Chenjie Xia
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Jun Ying
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Hongfeng Ruan
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhenyu Shi
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Rui Xu
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Taotao Xu
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang, China
| | - Shuaijie Lv
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang, China
| | - Liang Fang
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhen Zou
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Huihui Xu
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Luwei Xiao
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang, China
| | - Ping-Er Wang
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| |
Collapse
|
34
|
Böker KO, Richter K, Jäckle K, Taheri S, Grunwald I, Borcherding K, von Byern J, Hartwig A, Wildemann B, Schilling AF, Lehmann W. Current State of Bone Adhesives-Necessities and Hurdles. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E3975. [PMID: 31801225 PMCID: PMC6926991 DOI: 10.3390/ma12233975] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/20/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023]
Abstract
The vision of gluing two bone fragments with biodegradable and biocompatible adhesives remains highly fascinating and attractive to orthopedic surgeons. Possibly shorter operation times, better stabilization, lower infection rates, and unnecessary removal make this approach very appealing. After 30 years of research in this field, the first adhesive systems are now appearing in scientific reports that may fulfill the comprehensive requirements of bioadhesives for bone. For a successful introduction into clinical application, special requirements of the musculoskeletal system, challenges in the production of a bone adhesive, as well as regulatory hurdles still need to be overcome. In this article, we will give an overview of existing synthetic polymers, biomimetic, and bio-based adhesive approaches, review the regulatory hurdles they face, and discuss perspectives of how bone adhesives could be efficiently introduced into clinical application, including legal regulations.
Collapse
Affiliation(s)
- Kai O. Böker
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Goettingen, Robert Koch Straße 40, 37075 Göttingen, Germany; (K.J.); (S.T.); (A.F.S.); (W.L.)
| | - Katharina Richter
- Fraunhofer Institute for Manufacturing Technology and Advanced Materials (IFAM), Wiener Straße 12, 28359 Bremen, Germany; (K.R.); (K.B.); (A.H.)
| | - Katharina Jäckle
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Goettingen, Robert Koch Straße 40, 37075 Göttingen, Germany; (K.J.); (S.T.); (A.F.S.); (W.L.)
| | - Shahed Taheri
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Goettingen, Robert Koch Straße 40, 37075 Göttingen, Germany; (K.J.); (S.T.); (A.F.S.); (W.L.)
| | - Ingo Grunwald
- Industrial and Environmental Biology, Hochschule Bremen—City University of Applied Sciences, Neustadtswall 30, 28199 Bremen, Germany;
| | - Kai Borcherding
- Fraunhofer Institute for Manufacturing Technology and Advanced Materials (IFAM), Wiener Straße 12, 28359 Bremen, Germany; (K.R.); (K.B.); (A.H.)
| | - Janek von Byern
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, 1200 Vienna, Austria;
- Faculty of Life Science, University of Vienna, Core Facility Cell Imaging and Ultrastructure Research, Althanstrasse 14, 1090 Vienna, Austria
| | - Andreas Hartwig
- Fraunhofer Institute for Manufacturing Technology and Advanced Materials (IFAM), Wiener Straße 12, 28359 Bremen, Germany; (K.R.); (K.B.); (A.H.)
- Department 2 Biology/Chemistry, University of Bremen, Leobener Straße 3, 28359 Bremen, Germany
| | - Britt Wildemann
- Experimental Trauma Surgery, University Hospital Jena, 07747 Jena, Germany;
| | - Arndt F. Schilling
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Goettingen, Robert Koch Straße 40, 37075 Göttingen, Germany; (K.J.); (S.T.); (A.F.S.); (W.L.)
| | - Wolfgang Lehmann
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Goettingen, Robert Koch Straße 40, 37075 Göttingen, Germany; (K.J.); (S.T.); (A.F.S.); (W.L.)
| |
Collapse
|
35
|
Stefanowski J, Lang A, Rauch A, Aulich L, Köhler M, Fiedler AF, Buttgereit F, Schmidt-Bleek K, Duda GN, Gaber T, Niesner RA, Hauser AE. Spatial Distribution of Macrophages During Callus Formation and Maturation Reveals Close Crosstalk Between Macrophages and Newly Forming Vessels. Front Immunol 2019; 10:2588. [PMID: 31956322 PMCID: PMC6953593 DOI: 10.3389/fimmu.2019.02588] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/18/2019] [Indexed: 01/15/2023] Open
Abstract
Macrophages are essential players in the process of fracture healing, acting by remodeling of the extracellular matrix and enabling vascularization. Whilst activated macrophages of M1-like phenotype are present in the initial pro-inflammatory phase of hours to days of fracture healing, an anti-inflammatory M2-like macrophage phenotype is supposed to be crucial for the induction of downstream cascades of healing, especially the initiation of vascularization. In a mouse-osteotomy model, we provide a comprehensive characterization of vessel (CD31+, Emcn+) and macrophage phenotypes (F4/80, CD206, CD80, Mac-2) during the process of fracture healing. To this end, we phenotype the phases of vascular regeneration-the expansion phase (d1-d7 after injury) and the remodeling phase of the endothelial network, until tissue integrity is restored (d14-d21 after injury). Vessels which appear during the bone formation process resemble type H endothelium (CD31hiEmcnhi), and are closely connected to osteoprogenitors (Runx2+, Osx+) and F4/80+ macrophages. M1-like macrophages are present in the initial phase of vascularization until day 3 post osteotomy, but they are rare during later regeneration phases. M2-like macrophages localize mainly extramedullary, and CD206+ macrophages are found to express Mac-2+ during the expansion phase. VEGFA expression is initiated by CD80+ cells, including F4/80+ macrophages, until day 3, while subsequently osteoblasts and chondrocytes are main contributors to VEGFA production at the fracture site. Using Longitudinal Intravital Microendoscopy of the Bone (LIMB) we observe changes in the motility and organization of CX3CR1+ cells, which infiltrate the injury site after an osteotomy. A transient accumulation, resulting in spatial polarization of both, endothelial cells and macrophages, in regions distal to the fracture site, is evident. Immunofluorescence histology followed by histocytometric analysis reveals that F4/80+CX3CR1+ myeloid cells precede vascularization.
Collapse
Affiliation(s)
- Jonathan Stefanowski
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Annemarie Lang
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ariana Rauch
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Linus Aulich
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Markus Köhler
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Alexander F Fiedler
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Georg N Duda
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Raluca A Niesner
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Dynamic and Functional in vivo Imaging, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW This article reviews the past 2 years of research on Notch signaling as it relates to bone physiology, with the goal of reconciling seemingly discrepant findings and identifying fruitful areas of potential future research. RECENT FINDINGS Conditional animal models and high-throughput omics have contributed to a greater understanding of the context-dependent role of Notch signaling in bone. However, significant gaps remain in our understanding of how spatiotemporal context and epigenetic state dictate downstream Notch phenotypes. Biphasic activation of Notch signaling orchestrates progression of mesenchymal progenitor cells through the osteoblast lineage, but there is a limited understanding of ligand- and receptor-specific functions. Paracrine Notch signaling through non-osteoblastic cell types contributes additional layers of complexity, and we anticipate impactful future work related to the integration of these cell types and signaling mechanisms.
Collapse
Affiliation(s)
- Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Pl, Ann Arbor, MI, 48872, USA.
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Pl, Ann Arbor, MI, 48872, USA
| |
Collapse
|
37
|
Hadjiargyrou M, Komatsu DE. The Therapeutic Potential of MicroRNAs as Orthobiologics for Skeletal Fractures. J Bone Miner Res 2019; 34:797-809. [PMID: 30866092 PMCID: PMC6536331 DOI: 10.1002/jbmr.3708] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/04/2019] [Accepted: 02/23/2019] [Indexed: 12/19/2022]
Abstract
The repair of a fractured bone is critical to the well-being of humans. Failure of the repair process to proceed normally can lead to complicated fractures, exemplified by either a delay in union or a complete nonunion. Both of these conditions lead to pain, the possibility of additional surgery, and impairment of life quality. Additionally, work productivity decreases, income is reduced, and treatment costs increase, resulting in financial hardship. Thus, developing effective treatments for these difficult fractures or even accelerating the normal physiological repair process is warranted. Accumulating evidence shows that microRNAs (miRNAs), small noncoding RNAs, can serve as key regulatory molecules of fracture repair. In this review, a brief description of the fracture repair process and miRNA biogenesis is presented, as well as a summary of our current knowledge of the involvement of miRNAs in physiological fracture repair, osteoporotic fractures, and bone defect healing. Further, miRNA polymorphisms associated with fractures, miRNA presence in exosomes, and miRNAs as potential therapeutic orthobiologics are also discussed. This is a timely review as several miRNA-based therapeutics have recently entered clinical trials for nonskeletal applications and thus it is incumbent upon bone researchers to explore whether miRNAs can become the next class of orthobiologics for the treatment of skeletal fractures.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY 11568-8000
| | - David E. Komatsu
- Department of Orthopaedics, Stony Brook University, Stony Brook, NY 11794-8181
| |
Collapse
|
38
|
Baker CE, Moore-Lotridge SN, Hysong AA, Posey SL, Robinette JP, Blum DM, Benvenuti MA, Cole HA, Egawa S, Okawa A, Saito M, McCarthy JR, Nyman JS, Yuasa M, Schoenecker JG. Bone Fracture Acute Phase Response-A Unifying Theory of Fracture Repair: Clinical and Scientific Implications. Clin Rev Bone Miner Metab 2018; 16:142-158. [PMID: 30930699 PMCID: PMC6404386 DOI: 10.1007/s12018-018-9256-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone fractures create five problems that must be resolved: bleeding, risk of infection, hypoxia, disproportionate strain, and inability to bear weight. There have been enormous advancements in our understanding of the molecular mechanisms that resolve these problems after fractures, and in best clinical practices of repairing fractures. We put forth a modern, comprehensive model of fracture repair that synthesizes the literature on the biology and biomechanics of fracture repair to address the primary problems of fractures. This updated model is a framework for both fracture management and future studies aimed at understanding and treating this complex process. This model is based upon the fracture acute phase response (APR), which encompasses the molecular mechanisms that respond to injury. The APR is divided into sequential stages of "survival" and "repair." Early in convalescence, during "survival," bleeding and infection are resolved by collaborative efforts of the hemostatic and inflammatory pathways. Later, in "repair," avascular and biomechanically insufficient bone is replaced by a variable combination of intramembranous and endochondral ossification. Progression to repair cannot occur until survival has been ensured. A disproportionate APR-either insufficient or exuberant-leads to complications of survival (hemorrhage, thrombosis, systemic inflammatory response syndrome, infection, death) and/or repair (delayed- or non-union). The type of ossification utilized for fracture repair is dependent on the relative amounts of strain and vascularity in the fracture microenvironment, but any failure along this process can disrupt or delay fracture healing and result in a similar non-union. Therefore, incomplete understanding of the principles herein can result in mismanagement of fracture care or application of hardware that interferes with fracture repair. This unifying model of fracture repair not only informs clinicians how their interventions fit within the framework of normal biological healing but also instructs investigators about the critical variables and outputs to assess during a study of fracture repair.
Collapse
Affiliation(s)
- Courtney E Baker
- 1Department of Orthopaedics, Mayo Clinic, 200 1st Ave SW, Rochester, MN 55903 USA
| | - Stephanie N Moore-Lotridge
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,3Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Ave. South, Nashville, TN 37232 USA
| | - Alexander A Hysong
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - Samuel L Posey
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - J Patton Robinette
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - Deke M Blum
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - Michael A Benvenuti
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA
| | - Heather A Cole
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA
| | - Satoru Egawa
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Atsushi Okawa
- 5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Masanori Saito
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Jason R McCarthy
- Masonic Research Institute, 2150 Bleecker St, Utica, NY 13501 USA
| | - Jeffry S Nyman
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,7Department of Biomedical Engineering, Vanderbilt University, PMB 351631, 2301 Vanderbilt Place, Nashville, TN 37235 USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, F-519 VA Acre Building, 1210 24th Ave. South, Nashville, TN 37232 USA
| | - Masato Yuasa
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Jonathan G Schoenecker
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,3Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Ave. South, Nashville, TN 37232 USA.,9Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave, Robinson Research Building, Nashville, TN 37232 USA.,10Department of Pediatrics, Vanderbilt University Medical Center, 4202 Doctor's Office Tower, 2200 Children's Way, Nashville, TN 37232 USA
| |
Collapse
|