1
|
Maemura M, Morita M, Ogata S, Miyamoto Y, Ida T, Shibusaka K, Negishi S, Hosonuma M, Saito T, Yoshitake J, Takata T, Matsunaga T, Mishima E, Barayeu U, Akaike T, Yano F. Supersulfides contribute to joint homeostasis and bone regeneration. Redox Biol 2025; 81:103545. [PMID: 39983344 PMCID: PMC11893308 DOI: 10.1016/j.redox.2025.103545] [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: 12/04/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
The physiological functions of supersulfides, inorganic and organic sulfides with sulfur catenation, have been extensively studied. Their synthesis is mainly mediated by mitochondrial cysteinyl-tRNA synthetase (CARS2) that functions as a principal cysteine persulfide synthase. This study aimed to investigate the role of supersulfides in joint homeostasis and bone regeneration. Using Cars2AINK/+ mutant mice, in which the KIIK motif of CARS2 essential for supersulfide production was replaced with AINK, we evaluated the role of supersulfides in fracture healing and cartilage homeostasis during osteoarthritis (OA). Tibial fracture surgery was performed on the wild-type (Cars2+/+) and Cars2AINK/+ mice littermates. Bulk RNA-seq analysis for the osteochondral regeneration in the fracture model showed increased inflammatory markers and reduced osteogenic factors, indicative of impaired bone regeneration, in Cars2AINK/+ mice. Destabilization of the medial meniscus (DMM) surgery was performed to produce the mouse OA model. Histological analyses with Osteoarthritis Research Society International and synovitis scores revealed accelerated OA progression in Cars2AINK/+ mice compared with that in Cars2+/+ mice. To assess the effects of supersulfides on OA progression, glutathione trisulfide (GSSSG) or saline was periodically injected into the mouse knee joints after the DMM surgery. Thus, supersulfides derived from CARS2 and GSSSG exogenously administered significantly inhibited inflammation and lipid peroxidation of the joint cartilage, possibly through suppression of ferroptosis, during OA development. This study represents a significant advancement in understanding anti-inflammatory and anti-oxidant functions of supersulfides in skeletal tissues and may have a clinical relevance for the bone healing and OA therapeutics.
Collapse
Affiliation(s)
- Miki Maemura
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Seiryo Ogata
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Yoichi Miyamoto
- Faculty of Arts and Sciences at Fujiyoshida, Showa University, Fujiyoshida, Japan
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Kazuhiro Shibusaka
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan; Department of Orthodontics, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Soichiro Negishi
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Masahiro Hosonuma
- Department of Pharmacology, Graduate School of Pharmacy, Showa University, Tokyo, Japan
| | - Taku Saito
- Sensory & Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Yoshitake
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Tsuyoshi Takata
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan
| | - Tetsuro Matsunaga
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan; Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | | | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sedai, Japan.
| | - Fumiko Yano
- Department of Biochemistry, Graduate School of Dentistry, Showa University, Tokyo, Japan.
| |
Collapse
|
2
|
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
|
3
|
Trompet D, Melis S, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone development and repair. J Bone Miner Res 2024; 39:633-654. [PMID: 38696703 DOI: 10.1093/jbmr/zjae069] [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: 12/04/2023] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/04/2024]
Abstract
Bone development, growth, and repair are complex processes involving various cell types and interactions, with central roles played by skeletal stem and progenitor cells. Recent research brought new insights into the skeletal precursor populations that mediate intramembranous and endochondral bone development. Later in life, many of the cellular and molecular mechanisms determining development are reactivated upon fracture, with powerful trauma-induced signaling cues triggering a variety of postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect. Interestingly, in this injury context, the current evidence suggests that the fates of both SSPCs and differentiated skeletal cells can be considerably flexible and dynamic, and that multiple cell sources can be activated to operate as functional progenitors generating chondrocytes and/or osteoblasts. The combined implementation of in vivo lineage tracing, cell surface marker-based cell selection, single-cell molecular analyses, and high-resolution in situ imaging has strongly improved our insights into the diversity and roles of developmental and reparative stem/progenitor subsets, while also unveiling the complexity of their dynamics, hierarchies, and relationships. Albeit incompletely understood at present, findings supporting lineage flexibility and possibly plasticity among sources of osteogenic cells challenge the classical dogma of a single primitive, self-renewing, multipotent stem cell driving bone tissue formation and regeneration from the apex of a hierarchical and strictly unidirectional differentiation tree. We here review the state of the field and the newest discoveries in the origin, identity, and fates of skeletal progenitor cells during bone development and growth, discuss the contributions of adult SSPC populations to fracture repair, and reflect on the dynamism and relationships among skeletal precursors and differentiated cell lineages. Further research directed at unraveling the heterogeneity and capacities of SSPCs, as well as the regulatory cues determining their fate and functioning, will offer vital new options for clinical translation toward compromised fracture healing and bone regenerative medicine.
Collapse
Affiliation(s)
- Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Andrei S Chagin
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
4
|
Liu YL, Tang XT, Shu HS, Zou W, Zhou BO. Fibrous periosteum repairs bone fracture and maintains the healed bone throughout mouse adulthood. Dev Cell 2024; 59:1192-1209.e6. [PMID: 38554700 DOI: 10.1016/j.devcel.2024.03.019] [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: 04/02/2023] [Revised: 12/07/2023] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
Bone is regarded as one of few tissues that heals without fibrous scar. The outer layer of the periosteum is covered with fibrous tissue, whose function in bone formation is unknown. We herein developed a system to distinguish the fate of fibrous-layer periosteal cells (FL-PCs) from the skeletal stem/progenitor cells (SSPCs) in the cambium-layer periosteum and bone marrow in mice. We showed that FL-PCs did not participate in steady-state osteogenesis, but formed the main body of fibrocartilaginous callus during fracture healing. Moreover, FL-PCs invaded the cambium-layer periosteum and bone marrow after fracture, forming neo-SSPCs that continued to maintain the healed bones throughout adulthood. The FL-PC-derived neo-SSPCs expressed lower levels of osteogenic signature genes and displayed lower osteogenic differentiation activity than the preexisting SSPCs. Consistent with this, healed bones were thinner and formed more slowly than normal bones. Thus, the fibrous periosteum becomes the cellular origin of bones after fracture and alters bone properties permanently.
Collapse
Affiliation(s)
- Yiming Liam Liu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyu Thomas Tang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Sophie Shu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiguo Zou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Bo O Zhou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China.
| |
Collapse
|
5
|
Roberts JL, Kapfhamer D, Devarapalli V, Drissi H. IL-17RA Signaling in Prx1+ Mesenchymal Cells Influences Fracture Healing in Mice. Int J Mol Sci 2024; 25:3751. [PMID: 38612562 PMCID: PMC11011315 DOI: 10.3390/ijms25073751] [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: 02/10/2024] [Revised: 03/17/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Fracture healing is a complex series of events that requires a local inflammatory reaction to initiate the reparative process. This inflammatory reaction is important for stimulating the migration and proliferation of mesenchymal progenitor cells from the periosteum and surrounding tissues to form the cartilaginous and bony calluses. The proinflammatory cytokine interleukin (IL)-17 family has gained attention for its potential regenerative effects; however, the requirement of IL-17 signaling within mesenchymal progenitor cells for normal secondary fracture healing remains unknown. The conditional knockout of IL-17 receptor a (Il17ra) in mesenchymal progenitor cells was achieved by crossing Il17raF/F mice with Prx1-cre mice to generate Prx1-cre; Il17raF/F mice. At 3 months of age, mice underwent experimental unilateral mid-diaphyseal femoral fractures and healing was assessed by micro-computed tomography (µCT) and histomorphometric analyses. The effects of IL-17RA signaling on the osteogenic differentiation of fracture-activated periosteal cells was investigated in vitro. Examination of the intact skeleton revealed that the conditional knockout of Il17ra decreased the femoral cortical porosity but did not affect any femoral trabecular microarchitectural indices. After unilateral femoral fractures, Il17ra conditional knockout impacted the cartilage and bone composition of the fracture callus that was most evident early in the healing process (day 7 and 14 post-fracture). Furthermore, the in vitro treatment of fracture-activated periosteal cells with IL-17A inhibited osteogenesis. This study suggests that IL-17RA signaling within Prx1+ mesenchymal progenitor cells can influence the early stages of endochondral ossification during fracture healing.
Collapse
Affiliation(s)
- Joseph L. Roberts
- Department of Orthopaedics, Emory University, Atlanta, GA 30329, USA; (J.L.R.)
- Atlanta VA Health Care System, Decatur, GA 30033, USA
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - David Kapfhamer
- Department of Orthopaedics, Emory University, Atlanta, GA 30329, USA; (J.L.R.)
- Atlanta VA Health Care System, Decatur, GA 30033, USA
| | - Varsha Devarapalli
- Department of Orthopaedics, Emory University, Atlanta, GA 30329, USA; (J.L.R.)
- Atlanta VA Health Care System, Decatur, GA 30033, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University, Atlanta, GA 30329, USA; (J.L.R.)
- Atlanta VA Health Care System, Decatur, GA 30033, USA
| |
Collapse
|
6
|
Tan FH, Bronner ME. Regenerative loss in the animal kingdom as viewed from the mouse digit tip and heart. Dev Biol 2024; 507:44-63. [PMID: 38145727 PMCID: PMC10922877 DOI: 10.1016/j.ydbio.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
The myriad regenerative abilities across the animal kingdom have fascinated us for centuries. Recent advances in developmental, molecular, and cellular biology have allowed us to unearth a surprising diversity of mechanisms through which these processes occur. Developing an all-encompassing theory of animal regeneration has thus proved a complex endeavor. In this chapter, we frame the evolution and loss of animal regeneration within the broad developmental constraints that may physiologically inhibit regenerative ability across animal phylogeny. We then examine the mouse as a model of regeneration loss, specifically the experimental systems of the digit tip and heart. We discuss the digit tip and heart as a positionally-limited system of regeneration and a temporally-limited system of regeneration, respectively. We delve into the physiological processes involved in both forms of regeneration, and how each phase of the healing and regenerative process may be affected by various molecular signals, systemic changes, or microenvironmental cues. Lastly, we also discuss the various approaches and interventions used to induce or improve the regenerative response in both contexts, and the implications they have for our understanding regenerative ability more broadly.
Collapse
Affiliation(s)
- Fayth Hui Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
7
|
Wang X, Ge Q, Zeng Q, Zou K, Bao Z, Ying J, Wu Z, Jin H, Chen J, Xu T. Dnmt3b ablation affects fracture repair process by regulating apoptosis. BMC Musculoskelet Disord 2024; 25:180. [PMID: 38413962 PMCID: PMC10900613 DOI: 10.1186/s12891-024-07283-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/14/2024] [Indexed: 02/29/2024] Open
Abstract
PURPOSE Previous studies have shown that DNA methyltransferase 3b (Dnmt3b) is the only Dnmt responsive to fracture repair and Dnmt3b ablation in Prx1-positive stem cells and chondrocyte cells both delayed fracture repair. Our study aims to explore the influence of Dnmt3b ablation in Gli1-positive stem cells in fracture healing mice and the underlying mechanism. METHODS We generated Gli1-CreERT2; Dnmt3bflox/flox (Dnmt3bGli1ER) mice to operated tibia fracture. Fracture callus tissues of Dnmt3bGli1ER mice and control mice were collected and analyzed by X-ray, micro-CT, biomechanical testing, histopathology and TUNEL assay. RESULTS The cartilaginous callus significantly decrease in ablation of Dnmt3b in Gli1-positive stem cells during fracture repair. The chondrogenic and osteogenic indicators (Sox9 and Runx2) in the fracture healing tissues in Dnmt3bGli1ER mice much less than control mice. Dnmt3bGli1ER mice led to delayed bone callus remodeling and decreased biomechanical properties of the newly formed bone during fracture repair. Both the expressions of Caspase-3 and Caspase-8 were upregulated in Dnmt3bGli1ER mice as well as the expressions of BCL-2. CONCLUSIONS Our study provides an evidence that Dnmt3b ablation Gli1-positive stem cells can affect fracture healing and lead to poor fracture healing by regulating apoptosis to decrease chondrocyte hypertrophic maturation.
Collapse
Affiliation(s)
- Xu Wang
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qinwen Ge
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qinghe Zeng
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Kaiao Zou
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhengsheng Bao
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jun Ying
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhen Wu
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Hongting Jin
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| | - Jiali Chen
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| | - Taotao Xu
- Institute of Orthopedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
8
|
Zhang X, Deng C, Qi S. Periosteum Containing Implicit Stem Cells: A Progressive Source of Inspiration for Bone Tissue Regeneration. Int J Mol Sci 2024; 25:2162. [PMID: 38396834 PMCID: PMC10889827 DOI: 10.3390/ijms25042162] [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: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The periosteum is known as the thin connective tissue covering most bone surfaces. Its extrusive bone regeneration capacity was confirmed from the very first century-old studies. Recently, pluripotent stem cells in the periosteum with unique physiological properties were unveiled. Existing in dynamic contexts and regulated by complex molecular networks, periosteal stem cells emerge as having strong capabilities of proliferation and multipotential differentiation. Through continuous exploration of studies, we are now starting to acquire more insight into the great potential of the periosteum in bone formation and repair in situ or ectopically. It is undeniable that the periosteum is developing further into a more promising strategy to be harnessed in bone tissue regeneration. Here, we summarized the development and structure of the periosteum, cell markers, and the biological features of periosteal stem cells. Then, we reviewed their pivotal role in bone repair and the underlying molecular regulation. The understanding of periosteum-related cellular and molecular content will help enhance future research efforts and application transformation of the periosteum.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Chen Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Shengcai Qi
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| |
Collapse
|
9
|
Ye P, Gu R, Zhu H, Chen J, Han F, Nie X. SOX family transcription factors as therapeutic targets in wound healing: A comprehensive review. Int J Biol Macromol 2023; 253:127243. [PMID: 37806414 DOI: 10.1016/j.ijbiomac.2023.127243] [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: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
The SOX family plays a vital role in determining the fate of cells and has garnered attention in the fields of cancer research and regenerative medicine. It also shows promise in the study of wound healing, as it actively participates in the healing processes of various tissues such as skin, fractures, tendons, and the cornea. However, our understanding of the mechanisms behind the SOX family's involvement in wound healing is limited compared to its role in cancer. Gaining insight into its role, distribution, interaction with other factors, and modifications in traumatized tissues could provide valuable new knowledge about wound healing. Based on current research, SOX2, SOX7, and SOX9 are the most promising members of the SOX family for future interventions in wound healing. SOX2 and SOX9 promote the renewal of cells, while SOX7 enhances the microvascular environment. The SOX family holds significant potential for advancing wound healing research. This article provides a comprehensive review of the latest research advancements and therapeutic tools related to the SOX family in wound healing, as well as the potential benefits and challenges of targeting the SOX family for wound treatment.
Collapse
Affiliation(s)
- Penghui Ye
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Rifang Gu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China
| | - Huan Zhu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Jitao Chen
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
10
|
Xu Y, Zhuo J, Wang Q, Xu X, He M, Zhang L, Liu Y, Wu X, Luo K, Chen Y. Site-specific periosteal cells with distinct osteogenic and angiogenic characteristics. Clin Oral Investig 2023; 27:7437-7450. [PMID: 37848582 DOI: 10.1007/s00784-023-05333-3] [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: 06/28/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVES This study aimed to investigate the site-specific characteristics of rat mandible periosteal cells (MPCs) and tibia periosteal cells (TPCs) to assess the potential application of periosteal cells (PCs) in bone tissue engineering (BTE). MATERIALS AND METHODS MPCs and TPCs were isolated and characterized. The potential of proliferation, migration, osteogenesis and adipogenesis of MPCs and TPCs were evaluated by CCK-8, scratch assay, Transwell assay, alkaline phosphatase staining and activity, Alizarin Red S staining, RT‒qPCR, and Western blot (WB) assays, respectively. Then, these cells were cocultured with human umbilical vein endothelial cells (HUVECs) to investigate their angiogenic capacity, which was assessed by scratch assay, Transwell assay, Matrigel tube formation assay, RT‒qPCR, and WB assays. RESULTS MPCs exhibited higher osteogenic potential, higher alkaline phosphatase activity, and more mineralized nodule formation, while TPCs showed a greater capability for proliferation, migration, and adipogenesis. MPCs showed higher expression of angiogenic factors, and the conditioned medium of MPCs accelerated the migration of HUVECs, while MPC- conditioned medium induced the formation of more tubular structure in HUVECs in vitro. These data suggest that compared to TPCs, MPCs exert more consequential proangiogenic effects on HUVECs. CONCLUSIONS PCs possess skeletal site-specific differences in biological characteristics. MPCs exhibit more eminent osteogenic and angiogenic potentials, which highlights the potential application of MPCs for BTE. CLINICAL RELEVANCE Autologous bone grafting as the main modality for maxillofacial bone defect repair has many limitations. Constituting an important cell type in bone repair and regeneration, MPCs show greater potential for application in BTE, which provides a promising treatment option for maxillofacial bone defect repair.
Collapse
Affiliation(s)
- Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Jin Zhuo
- Xuzhou Stomatological Hospital, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Qisong Wang
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 354000, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Xiaohong Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| | - Yuling Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| |
Collapse
|
11
|
Steppe L, Megafu M, Tschaffon-Müller ME, Ignatius A, Haffner-Luntzer M. Fracture healing research: Recent insights. Bone Rep 2023; 19:101686. [PMID: 38163010 PMCID: PMC10757288 DOI: 10.1016/j.bonr.2023.101686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 01/03/2024] Open
Abstract
Bone has the rare capability of scarless regeneration that enables the complete restoration of the injured bone area. In recent decades, promising new technologies have emerged from basic, translational and clinical research for fracture treatment; however, 5-10 % of all bone fractures still fail to heal successfully or heal in a delayed manner. Several comorbidities and risk factors have been identified which impair bone healing and might lead to delayed bone union or non-union. Therefore, a considerable amount of research has been conducted to elucidate molecular mechanisms of successful and delayed fracture healing to gain further insights into this complex process. One focus of recent research is to investigate the complex interactions of different cell types and the action of progenitor cells during the healing process. Of particular interest is also the identification of patient-specific comorbidities and how these affect fracture healing. In this review, we discuss the recent knowledge about progenitor cells for long bone repair and the influence of comorbidities such as diabetes, postmenopausal osteoporosis, and chronic stress on the healing process. The topic selection for this review was made based on the presented studies at the 2022 annual meeting of the European Calcified Tissue Society (ECTS) in Helsinki.
Collapse
Affiliation(s)
- Lena Steppe
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Germany
| | - Michael Megafu
- A.T. Still University Kirksville College of Osteopathic Medicine, USA
| | | | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Germany
| | | |
Collapse
|
12
|
Lee KK, Changoor A, Grynpas MD, Mitchell J. Increased Osteoblast Gα S Promotes Ossification by Suppressing Cartilage and Enhancing Callus Mineralization During Fracture Repair in Mice. JBMR Plus 2023; 7:e10841. [PMID: 38130768 PMCID: PMC10731140 DOI: 10.1002/jbm4.10841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 12/23/2023] Open
Abstract
GαS, the stimulatory G protein α-subunit that raises intracellular cAMP levels by activating adenylyl cyclase, plays a vital role in bone development, maintenance, and remodeling. Previously, using transgenic mice overexpressing GαS in osteoblasts (GS-Tg), we demonstrated the influence of osteoblast GαS level on osteogenesis, bone turnover, and skeletal responses to hyperparathyroidism. To further investigate whether alterations in GαS levels affect endochondral bone repair, a postnatal bone regenerative process that recapitulates embryonic bone development, we performed stabilized tibial osteotomy in male GS-Tg mice at 8 weeks of age and examined the progression of fracture healing by micro-CT, histomorphometry, and gene expression analysis over a 4-week period. Bone fractures from GS-Tg mice exhibited diminished cartilage formation at the time of peak soft callus formation at 1 week post-fracture followed by significantly enhanced callus mineralization and new bone formation at 2 weeks post-fracture. The opposing effects on chondrogenesis and osteogenesis were validated by downregulation of chondrogenic markers and upregulation of osteogenic markers. Histomorphometric analysis at times of increased bone formation (2 and 3 weeks post-fracture) revealed excess fibroblast-like cells on newly formed woven bone surfaces and elevated osteocyte density in GS-Tg fractures. Coincident with enhanced callus mineralization and bone formation, GS-Tg mice showed elevated active β-catenin and Wntless proteins in osteoblasts at 2 weeks post-fracture, further substantiated by increased mRNA encoding various canonical Wnts and Wnt target genes, suggesting elevated osteoblastic Wnt secretion and Wnt/β-catenin signaling. The GS-Tg bony callus at 4 weeks post-fracture exhibited greater mineral density and decreased polar moment of inertia, resulting in improved material stiffness. These findings highlight that elevated GαS levels increase Wnt signaling, conferring an increased osteogenic differentiation potential at the expense of chondrogenic differentiation, resulting in improved mechanical integrity. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Kathy K Lee
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoCanada
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoCanada
| | - Adele Changoor
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoCanada
- Department of SurgeryUniversity of TorontoTorontoCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoCanada
| | - Marc D Grynpas
- Lunenfeld‐Tanenbaum Research InstituteMount Sinai HospitalTorontoCanada
- Department of SurgeryUniversity of TorontoTorontoCanada
| | - Jane Mitchell
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoCanada
| |
Collapse
|
13
|
Rifai A, Weerasinghe DK, Tilaye GA, Nisbet D, Hodge JM, Pasco JA, Williams LJ, Samarasinghe RM, Williams RJ. Biofabrication of functional bone tissue: defining tissue-engineered scaffolds from nature. Front Bioeng Biotechnol 2023; 11:1185841. [PMID: 37614632 PMCID: PMC10444209 DOI: 10.3389/fbioe.2023.1185841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
Damage to bone leads to pain and loss of movement in the musculoskeletal system. Although bone can regenerate, sometimes it is damaged beyond its innate capacity. Research interest is increasingly turning to tissue engineering (TE) processes to provide a clinical solution for bone defects. Despite the increasing biomimicry of tissue-engineered scaffolds, significant gaps remain in creating the complex bone substitutes, which include the biochemical and physical conditions required to recapitulate bone cells' natural growth, differentiation and maturation. Combining advanced biomaterials with new additive manufacturing technologies allows the development of 3D tissue, capable of forming cell aggregates and organoids based on natural and stimulated cues. Here, we provide an overview of the structure and mechanical properties of natural bone, the role of bone cells, the remodelling process, cytokines and signalling pathways, causes of bone defects and typical treatments and new TE strategies. We highlight processes of selecting biomaterials, cells and growth factors. Finally, we discuss innovative tissue-engineered models that have physiological and anatomical relevance for cancer treatments, injectable stimuli gels, and other therapeutic drug delivery systems. We also review current challenges and prospects of bone TE. Overall, this review serves as guide to understand and develop better tissue-engineered bone designs.
Collapse
Affiliation(s)
- Aaqil Rifai
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - D. Kavindi Weerasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Gebreselassie Addisu Tilaye
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - David Nisbet
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Laboratory of Advanced Biomaterials, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| | - Jason M. Hodge
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Julie A. Pasco
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Medicine-Western Health, The University of Melbourne, St Albans, VIC, Australia
| | - Lana J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Rasika M. Samarasinghe
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard J. Williams
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC, Australia
- Aikenhead Centre for Medical Discovery, St. Vincent’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Jun JY, Kim JH, Kim M, Hong S, Kim M, Ryu GH, Park JH, Jung HS, Sohn Y. Persicae Semen Promotes Bone Union in Rat Fractures by Stimulating Osteoblastogenesis through BMP-2 and Wnt Signaling. Int J Mol Sci 2023; 24:ijms24087388. [PMID: 37108563 PMCID: PMC10138545 DOI: 10.3390/ijms24087388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Fractures cause extreme pain to patients and impair movement, thereby significantly reducing their quality of life. However, in fracture patients, movement of the fracture site is restricted through application of a cast, and they are reliant on conservative treatment through calcium intake. Persicae semen (PS) is the dried mature seeds of Prunus persica (L.) Batsch, and in this study the effects of PS on osteoblast differentiation and bone union promotion were investigated. The osteoblast-differentiation-promoting effect of PS was investigated through alizarin red S and Von Kossa staining, and the regulatory role of PS on BMP-2 (Bmp2) and Wnt (Wnt10b) signaling, representing a key mechanism, was demonstrated at the protein and mRNA levels. In addition, the bone-union-promoting effect of PS was investigated in rats with fractured femurs. The results of the cell experiments showed that PS promotes mineralization and upregulates RUNX2 through BMP-2 and Wnt signaling. PS induced the expression of various osteoblast genes, including Alpl, Bglap, and Ibsp. The results of animal experiments show that the PS group had improved bone union and upregulated expression of osteogenic genes. Overall, the results of this study suggest that PS can promote fracture recovery by upregulating osteoblast differentiation and bone formation, and thus can be considered a new therapeutic alternative for fracture patients.
Collapse
Affiliation(s)
- Jae-Yun Jun
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| | - Jae-Hyun Kim
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| | - Minsun Kim
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| | - Sooyeon Hong
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| | - Myunghyun Kim
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| | - Gwang-Hyun Ryu
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| | - Jae Ho Park
- Department of Pharmaceutical Science, Jungwon University, Goesan-eup 28024, Republic of Korea
| | - Hyuk-Sang Jung
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| | - Youngjoo Sohn
- Department of Anatomy, College of Korean Medicine, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
Xiao H, Yan A, Li M, Wang L, Xiang J. LIPUS accelerates bone regeneration via HDAC6-mediated ciliogenesis. Biochem Biophys Res Commun 2023; 641:34-41. [PMID: 36521283 DOI: 10.1016/j.bbrc.2022.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022]
Abstract
Delayed fracture union and nonunion are common complications of fracture encountered, while Low-intensity pulsed ultrasound (LIPUS) can stimulate bone regeneration. Still, the underlying mechanism of LIPUS on bone regeneration has been poorly understood, which resulted in varied outcomes in the clinic. Therefore, figuring out the mechanism of LIPUS on bone regeneration can lay the foundation for better use of LIPUS in clinical bone regenerative therapies. In this study, we created transgenic mice to reveal the relationship between the periosteal cells' fate and the number of ciliated cells under the LIPUS stimulation. In vitro, we isolated the periosteal cell and aim to figure out the relationship between LIPUS and HDAC6-mediated ciliogenesis and find out a potential target for LIPUS-based bone regeneration strategies. The results showed that LIPUS promoted femoral bone defect regeneration and enhanced osteogenic differentiation of Prrx1+ cells. However, these pro-effects were significantly weakened when the Prrx1+ cell's primary cilia were knocked down. Besides, LIPUS stimulated the formation of Prrx1+ cells' primary cilia in the bone defect microenvironment. In vitro, the results supported that LIPUS enhanced the osteogenic differentiation of Prrx1+ cells through HDAC6-mediated ciliogenesis. In conclusion, λ LIPUS could promote the osteogenic differentiation of Prrx1+ cells to stimulate bone regeneration and inhibit the expression of HDAC6 to increase the prevalence of primary cilia in Prrx1+ cells. LIPUS could enhance the osteogenic differentiation of Prrx1+ cells mainly through HDAC6-mediated ciliogenesis.
Collapse
Affiliation(s)
- Han Xiao
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, Changsha, Hunan, 410007, China; The Pediatric Academy of University of South China, Changsha, Hunan, 410007, China
| | - An Yan
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, Changsha, Hunan, 410007, China; The Pediatric Academy of University of South China, Changsha, Hunan, 410007, China
| | - Miao Li
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, Changsha, Hunan, 410007, China; The Pediatric Academy of University of South China, Changsha, Hunan, 410007, China
| | - Linfeng Wang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jie Xiang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, 516600, China.
| |
Collapse
|
16
|
Nakayama M, Okada H, Seki M, Suzuki Y, Chung UI, Ohba S, Hojo H. Single-cell RNA sequencing unravels heterogeneity of skeletal progenitors and cell-cell interactions underlying the bone repair process. Regen Ther 2022; 21:9-18. [PMID: 35619947 PMCID: PMC9127115 DOI: 10.1016/j.reth.2022.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/10/2022] [Accepted: 05/03/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Activation of skeletal progenitors upon tissue injury and the subsequent cell fate specification are tightly coordinated in the bone repair process. Although known osteoimmunological signaling networks play important roles in the microenvironment of the bone defect sites, the molecular mechanism underlying the bone repair process has not been fully understood. Methods To better understand the behavior of the skeletal progenitors and the heterogeneity of the cells during bone repair at the microenvironmental level, we performed a combinatorial analysis consisting of lineage tracing for skeletal progenitors using the Sox9-CreERT2;R26R tdTomato mouse line followed by single-cell RNA sequencing (scRNA-seq) analysis using a mouse model of calvarial bone repair. To identify a therapeutic target for bone regeneration, further computational analysis was performed focusing on the identification of the cell-cell interactions, followed by pharmacological assessments with a critical-size calvarial bone defect mouse model. Results Lineage tracing analysis showed that skeletal progenitors marked by Sox9 were activated upon bone injury and contributed to bone repair by differentiating into osteoblasts. The scRNA-seq analysis characterized heterogeneous cell populations at the bone defect sites; the computational analysis predicted a bifurcated lineage from skeletal progenitors toward osteogenic and adipogenic lineages. Chemokine C-C motif ligand 9 (Ccl9) was identified as a signaling molecule that regulates bone regeneration in the mouse model, possibly through the regulation of adipogenic differentiation at the bone defect site. Conclusion Multipotential skeletal progenitors and the direction of the cell differentiation were characterized at single cell resolution in a mouse bone repair model. The Ccl9 signaling pathway may be a key factor directing osteogenesis from the progenitors in the model and may be a therapeutic target for bone regeneration.
Collapse
Affiliation(s)
- Mika Nakayama
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Hiroyuki Okada
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Ung-il Chung
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, 113-8655, Japan
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Hironori Hojo
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, 113-8655, Japan
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| |
Collapse
|
17
|
Jeffery EC, Mann TLA, Pool JA, Zhao Z, Morrison SJ. Bone marrow and periosteal skeletal stem/progenitor cells make distinct contributions to bone maintenance and repair. Cell Stem Cell 2022; 29:1547-1561.e6. [PMID: 36272401 DOI: 10.1016/j.stem.2022.10.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/15/2022] [Accepted: 10/06/2022] [Indexed: 01/09/2023]
Abstract
A fundamental question in bone biology concerns the contributions of skeletal stem/progenitor cells (SSCs) in the bone marrow versus the periosteum to bone repair. We found that SSCs in adult bone marrow can be identified based on Leprcre and Adiponectin-cre/creER expression while SSCs in adult periosteum can be identified based on Gli1creERT2 expression. Under steady-state conditions, new bone arose primarily from bone marrow SSCs. After bone injuries, both SSC populations began proliferating but made very different contributions to bone repair. Drill injuries were primarily repaired by LepR+/Adiponectin+ bone marrow SSCs. Conversely, bicortical fractures were primarily repaired by Gli1+ periosteal SSCs, though LepR+/Adiponectin+ bone marrow cells transiently formed trabecular bone at the fracture site. Gli1+ periosteal cells also regenerated LepR+ bone marrow stromal cells that expressed hematopoietic niche factors at fracture sites. Different bone injuries are thus repaired by different SSCs, with periosteal cells regenerating bone and marrow stroma after non-stabilized fractures.
Collapse
Affiliation(s)
- Elise C Jeffery
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Terry L A Mann
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jade A Pool
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J Morrison
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The periosteum, the outer layer of bone, is a major source of skeletal stem/progenitor cells (SSPCs) for bone repair. Here, we discuss recent findings on the characterization, role, and regulation of periosteal SSPCs (pSSPCs) during bone regeneration. RECENT FINDINGS Several markers have been described for pSSPCs but lack tissue specificity. In vivo lineage tracing and transcriptomic analyses have improved our understanding of pSSPC functions during bone regeneration. Bone injury activates pSSPCs that migrate, proliferate, and have the unique potential to form both bone and cartilage. The injury response of pSSPCs is controlled by many signaling pathways including BMP, FGF, Notch, and Wnt, their metabolic state, and their interactions with the blood clot, nerve fibers, blood vessels, and macrophages in the fracture environment. Periosteal SSPCs are essential for bone regeneration. Despite recent advances, further studies are required to elucidate pSSPC heterogeneity and plasticity that make them a central component of the fracture healing process and a prime target for clinical applications.
Collapse
Affiliation(s)
- Simon Perrin
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Creteil, France
| | - Céline Colnot
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Creteil, France.
| |
Collapse
|
19
|
Tang Y, Wu B, Huang T, Wang H, Shi R, Lai W, Xiang L. Collision of Commonality and Personalization: Better Understanding of the Periosteum. TISSUE ENGINEERING PART B: REVIEWS 2022; 29:91-102. [PMID: 36006374 DOI: 10.1089/ten.teb.2022.0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The periosteum is quite essential for bone repair. The excellent osteogenic properties of periosteal tissue make it a popular choice for accelerated osteogenesis in tissue engineering. With advances in research and technology, renewed attention has been paid to the periosteum. Recent studies have shown that the complexity of the periosteum is not only limited to histological features but also includes genetic and phenotypic features. In addition, the periosteum is proved to be quite site-specific in many ways. This brings challenges to the selection of periosteal donor sites. Limited understanding of the periosteum sets up barriers to developing optimal tissue regeneration strategies. A better understanding of periosteum could lead to better applications. Therefore, we reviewed the histological structure, gene expression, and function of the periosteum from both the commonality and personalization. It aims to discuss some obscure issues and untapped potential of periosteum and artificial periosteum in the application, where further theoretical research is needed. Overall, the site-specificity of the periosteum needs to be fully considered in future applications. However, significant further work is needed in relevant clinical trials to promote the further development of artificial periosteum.
Collapse
Affiliation(s)
- Yufei Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthdontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China,
| | - Bingfeng Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China,
| | - Tianyu Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China,
| | - Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China,
| | - Ruijianghan Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China,
| | - Wenli Lai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthdontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China,
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, No 14th, 3rd section, Renmin South Road, Chengdu, 610041, China, Chengdu, Sichuan Province, China, 610041,
| |
Collapse
|
20
|
Mudigonda S, Shah S, Das N, Corpuz JM, Ninkovic N, Al-Jezani N, Underhill TM, Salo PT, Mitha AP, Lyons FG, Cho R, Schmidt TA, Dufour A, Krawetz RJ. Proteoglycan 4 is present within the dura mater and produced by mesenchymal progenitor cells. Cell Tissue Res 2022; 389:483-499. [PMID: 35704103 DOI: 10.1007/s00441-022-03647-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/02/2022] [Indexed: 11/25/2022]
Abstract
Mesenchymal progenitor cells (MPCs) have been recently identified in human and murine epidural fat and have been hypothesized to contribute to the maintenance/repair/regeneration of the dura mater. MPCs can secrete proteoglycan 4 (PRG4/lubricin), and this protein can regulate tissue homeostasis through bio-lubrication and immunomodulatory functions. MPC lineage tracing reporter mice (Hic1) and human epidural fat MPCs were used to determine if PRG4 is expressed by these cells in vivo. PRG4 expression co-localized with Hic1+ MPCs in the dura throughout skeletal maturity and was localized adjacent to sites of dural injury. When Hic1+ MPCs were ablated, PRG4 expression was retained in the dura, yet when Prx1+ MPCs were ablated, PRG4 expression was completely lost. A number of cellular processes were impacted in human epidural fat MPCs treated with rhPRG4, and human MPCs contributed to the formation of epidural fat, and dura tissues were xenotransplanted into mouse dural injuries. We have shown that human and mouse MPCs in the epidural/dura microenvironment produce PRG4 and can contribute to dura homeostasis/repair/regeneration. Overall, these results suggest that these MPCs have biological significance within the dural microenvironment and that the role of PRG4 needs to be further elucidated.
Collapse
Affiliation(s)
- Sathvika Mudigonda
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Sophia Shah
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Nabangshu Das
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Jessica May Corpuz
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Nicoletta Ninkovic
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Nedaa Al-Jezani
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - T Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Paul T Salo
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alim P Mitha
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Frank G Lyons
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Roger Cho
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, CT, USA
| | - Antoine Dufour
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Roman J Krawetz
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada. .,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada. .,Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
21
|
Liu J, Zhang J, Lin X, Boyce BF, Zhang H, Xing L. Age-associated callus senescent cells produce TGF-β1 that inhibits fracture healing in aged mice. J Clin Invest 2022; 132:e148073. [PMID: 35426372 PMCID: PMC9012290 DOI: 10.1172/jci148073] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 02/16/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence plays an important role in human diseases, including osteoporosis and osteoarthritis. Senescent cells (SCs) produce the senescence-associated secretory phenotype to affect the function of neighboring cells and SCs themselves. Delayed fracture healing is common in the elderly and is accompanied by reduced mesenchymal progenitor cells (MPCs). However, the contribution of cellular senescence to fracture healing in the aged has not to our knowledge been studied. Here, we used C57BL/6J 4-month-old young and 20-month-old aged mice and demonstrated a rapid increase in SCs in the fracture callus of aged mice. The senolytic drugs dasatinib plus quercetin enhanced fracture healing in aged mice. Aged callus SCs inhibited the growth and proliferation of callus-derived MPCs (CaMPCs) and expressed high levels of TGF-β1. TGF-β-neutralizing Ab prevented the inhibitory effects of aged callus SCs on CaMPCs and promoted fracture healing in aged mice, which was associated with increased CaMPCs and proliferating cells. Thus, fracture triggered a significant cellular senescence in the callus cells of aged mice, which inhibited MPCs by expressing TGF-β1. Short-term administration of dasatinib plus quercetin depleted callus SCs and accelerated fracture healing in aged mice. Senolytic drugs represent a promising therapy, while TGF-β1 signaling is a molecular mechanism for fractures in the elderly via SCs.
Collapse
Affiliation(s)
- Jiatong Liu
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Jun Zhang
- Plastic Surgery Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
22
|
Zalama E, Karrouf G, Rizk A, Salama B, Samy A. Does zinc oxide nanoparticles potentiate the regenerative effect of platelet-rich fibrin in healing of critical bone defect in rabbits? BMC Vet Res 2022; 18:130. [PMID: 35366880 PMCID: PMC8976312 DOI: 10.1186/s12917-022-03231-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/15/2022] [Indexed: 11/20/2022] Open
Abstract
Background Many encouraging studies confirmed the ability of Zinc Oxide Nanoparticles (ZnONPs) in accelerating bone growth and mineralization. The use of Platelet Rich-Fibrin (PRF) as a sole filling material for large segmental bone defects remains questionable. The objectives are to investigate the regenerative efficacy of autologous Platelet Rich-Fibrin (PRF) and Zinc Oxide Nanoparticles (ZnONPs) in repairing large segmental bone ulnar defects in a randomized controlled study in rabbits using computed tomographic interpretations. A 12 mm critical size defect was surgically induced in the ulna of 30 rabbits (n = 10/ group). In the control group, the defect was left empty. In the PRF group, the defect is filled with PRF. In the PRF/ZnONPs group, the defect is filled with PRF that was inoculated with 0.1 ml of 0.2% ZnONPs. Radiologic healing capacity was evaluated at the first, second, and third postoperative months. Results Statistical analysis showed significant differences in the radiologic healing scores between the groups (P = 0.000–0.0001) at all-time points (P = 0.000–0.047) during the study. Conclusion Rabbits in the PRF/ZnONPs group showed the highest appreciable bone quality and quantity followed by the PRF group with high quantity but low bone quality meanwhile, rabbits in the control group showed minimal quantity but medium bone quality. Interestingly, the addition of ZnONPs to PRF can accelerate the healing of ulnar critical-size defects in rabbits.
Collapse
|
23
|
On the horizon: Hedgehog signaling to heal broken bones. Bone Res 2022; 10:13. [PMID: 35165260 PMCID: PMC8844053 DOI: 10.1038/s41413-021-00184-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022] Open
Abstract
Uncovering the molecular pathways that drive skeletal repair has been an ongoing challenge. Initial efforts have relied on in vitro assays to identify the key signaling pathways that drive cartilage and bone differentiation. While these assays can provide some clues, assessing specific pathways in animal models is critical. Furthermore, definitive proof that a pathway is required for skeletal repair is best provided using genetic tests. Stimulating the Hh (Hedgehog) pathway can promote cartilage and bone differentiation in cell culture assays. In addition, the application of HH protein or various pathway agonists in vivo has a positive influence on bone healing. Until recently, however, genetic proof that the Hh pathway is involved in bone repair has been lacking. Here, we consider both in vitro and in vivo studies that examine the role of Hh in repair and discuss some of the challenges inherent in their interpretation. We also identify needed areas of study considering a new appreciation for the role of cartilage during repair, the variety of cell types that may have differing roles in repair, and the recent availability of powerful lineage tracing techniques. We are optimistic that emerging genetic tools will make it possible to precisely define when and in which cells promoting Hh signaling can best promote skeletal repair, and thus, the clinical potential for targeting the Hh pathway can be realized.
Collapse
|
24
|
Targeted activation of androgen receptor signaling in the periosteum improves bone fracture repair. Cell Death Dis 2022; 13:123. [PMID: 35136023 PMCID: PMC8826926 DOI: 10.1038/s41419-022-04595-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/30/2021] [Accepted: 01/27/2022] [Indexed: 12/03/2022]
Abstract
Low testosterone level is an independent predictor of osteoporotic fracture in elderly men as well as increased fracture risk in men undergoing androgen deprivation. Androgens and androgen receptor (AR) actions are essential for bone development and homeostasis but their linkage to fracture repair remains unclear. Here we found that AR is highly expressed in the periosteum cells and is co-localized with a mesenchymal progenitor cell marker, paired-related homeobox protein 1 (Prrx1), during bone fracture repair. Mice lacking the AR gene in the periosteum expressing Prrx1-cre (AR-/Y;Prrx1::Cre) but not in the chondrocytes (AR-/Y;Col-2::Cre) exhibits reduced callus size and new bone volume. Gene expression data analysis revealed that the expression of several collagens, integrins and cell adhesion molecules were downregulated in periosteum-derived progenitor cells (PDCs) from AR-/Y;Prrx1::Cre mice. Mechanistically, androgens-AR signaling activates the AR/ARA55/FAK complex and induces the collagen-integrin α2β1 gene expression that is required for promoting the AR-mediated PDCs migration. Using mouse cortical-defect and femoral graft transplantation models, we proved that elimination of AR in periosteum of host mice impairs fracture healing, regardless of AR existence of transplanted donor graft. While testosterone implanted scaffolds failed to complete callus bridging across the fracture gap in AR-/Y;Prrx1::Cre mice, cell-based transplantation using DPCs re-expressing AR could lead to rescue bone repair. In conclusion, targeting androgen/AR axis in the periosteum may provide a novel therapy approach to improve fracture healing.
Collapse
|
25
|
Dai K, Deng S, Yu Y, Zhu F, Wang J, Liu C. Construction of developmentally inspired periosteum-like tissue for bone regeneration. Bone Res 2022; 10:1. [PMID: 34975148 PMCID: PMC8720863 DOI: 10.1038/s41413-021-00166-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 05/19/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022] Open
Abstract
The periosteum, a highly vascularized thin tissue, has excellent osteogenic and bone regenerative abilities. The generation of periosteum-mimicking tissue has become a novel strategy for bone defect repair and regeneration, especially in critical-sized bone defects caused by trauma and bone tumor resection. Here, we utilized a bone morphogenetic protein-2 (BMP-2)-loaded scaffold to create periosteum-like tissue (PT) in vivo, mimicking the mesenchymal condensation during native long bone development. We found that BMP-2-induced endochondral ossification plays an indispensable role in the construction of PTs. Moreover, we confirmed that BMP-2-induced PTs exhibit a similar architecture to the periosteum and harbor abundant functional periosteum-like tissue-derived cells (PTDCs), blood vessels, and osteochondral progenitor cells. Interestingly, we found that the addition of chondroitin sulfate (CS), an essential component of the extracellular matrix (ECM), could further increase the abundance and enhance the function of recruited PTDCs from the PTs and finally increase the regenerative capacity of the PTs in autologous transplantation assays, even in old mice. This novel biomimetic strategy for generating PT through in vivo endochondral ossification deserves further clinical translation.
Collapse
Affiliation(s)
- Kai Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China.,Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, P. R. China
| | - Shunshu Deng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China.,Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, P. R. China
| | - Yuanman Yu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China.,Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, P. R. China
| | - Fuwei Zhu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China.,Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, P. R. China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China. .,Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, P. R. China.
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, P. R. China. .,Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, P. R. China. .,Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, P. R. China.
| |
Collapse
|
26
|
Shah S, Mudigonda S, Underhill TM, Salo PT, Mitha AP, Krawetz RJ. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:200-212. [PMID: 35259263 PMCID: PMC8929447 DOI: 10.1093/stcltm/szab014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/14/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Sophia Shah
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Sathvika Mudigonda
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Tully Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Paul T Salo
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alim P Mitha
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Roman J Krawetz
- Corresponding author: Roman J. Krawetz, McCaig Institute for Bone and Joint Health, University of Calgary, HRIC 3AA10, 3330 Hospital Dr NW, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Paul GR, Wehrle E, Tourolle DC, Kuhn GA, Müller R. Real-time finite element analysis allows homogenization of tissue scale strains and reduces variance in a mouse defect healing model. Sci Rep 2021; 11:13511. [PMID: 34188165 PMCID: PMC8241979 DOI: 10.1038/s41598-021-92961-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 06/18/2021] [Indexed: 11/26/2022] Open
Abstract
Mechanical loading allows both investigation into the mechano-regulation of fracture healing as well as interventions to improve fracture-healing outcomes such as delayed healing or non-unions. However, loading is seldom individualised or even targeted to an effective mechanical stimulus level within the bone tissue. In this study, we use micro-finite element analysis to demonstrate the result of using a constant loading assumption for all mouse femurs in a given group. We then contrast this with the application of an adaptive loading approach, denoted real time Finite Element adaptation, in which micro-computed tomography images provide the basis for micro-FE based simulations and the resulting strains are manipulated and targeted to a reference distribution. Using this approach, we demonstrate that individualised femoral loading leads to a better-specified strain distribution and lower variance in tissue mechanical stimulus across all mice, both longitudinally and cross-sectionally, while making sure that no overloading is occurring leading to refracture of the femur bones.
Collapse
Affiliation(s)
- Graeme R Paul
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Duncan C Tourolle
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland.
| |
Collapse
|
30
|
Van Camp N, Verhelst PJ, Nicot R, Ferri J, Politis C. Impaired Callus Formation in Pathological Mandibular Fractures in Medication-Related Osteonecrosis of the Jaw and Osteoradionecrosis. J Oral Maxillofac Surg 2021; 79:1892-1901. [PMID: 34097863 DOI: 10.1016/j.joms.2021.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Nonsurgical treatment of mandibular fractures secondary to medication-related osteonecrosis of the jaw (MRONJ) or osteoradionecrosis (ORN) mostly results in nonunion, whereas nonsurgical fracture treatment of atrophic fractures can achieve favorable results in selected cases. The aim of this study was to compare callus formation in pathological mandibular fractures due to MRONJ, ORN, or extreme mandibular atrophy. METHODS A retrospective cohort study reviewing the medical records of all MRONJ-, ORN-, or atrophy-related fractures treated at the departments of maxillofacial surgery in the Leuven or Lille university hospitals between 2010 and 2019 was undertaken. The primary predictor variable in this study was disease state (MRONJ, ORN, or extreme mandibular atrophy). The primary outcome variable was callus formation after 1 month of follow-up (present, absent). Additional study variables measured included patient age and gender. T-tests, Fisher exact tests, and multiple logistic regression were used for statistical analysis. The significance level was set at P < .05. RESULTS Seventy patients were analyzed (12 MRONJ cases, 54 ORN fractures, 4 atrophic fractures). The callus formation prevalence in nonsurgically approached fractures secondary to ORN and MRONJ after 1 month of follow-up was 3.03% (2/66 cases). In contrast, callus was detected in all patients in the mandibular atrophy-related fracture group. Osteonecrosis was statistically correlated with impaired callus formation (P = .0121). CONCLUSION Whereas one would expect indirect fracture healing and thus callus formation to occur in all non-surgically treated fractures, our data demonstrate its absence in the majority of MRONJ- and ORN-related fractures. Multiple plausible explanations for this phenomenon were identified: periosteal damage with loss of callus-forming cells, compromised vasculature, and bacterial colonization.
Collapse
Affiliation(s)
- Nathalie Van Camp
- Resident, Oral and Maxillofacial Surgery - Imaging and Pathology Research Group, Department of Imaging and Pathology, Faculty of Medicine, University of Leuven and Department of Oral & Maxillofacial Surgery, University of Leuven, Leuven, Belgium.
| | - Pieter-Jan Verhelst
- Resident, Oral and Maxillofacial Surgery - Imaging and Pathology Research Group, Department of Imaging and Pathology, Faculty of Medicine, University of Leuven and Department of Oral & Maxillofacial Surgery, University of Leuven, Leuven, Belgium
| | - Romain Nicot
- Staff Physician, Stomatologie, Chirurgie Orale et Maxillofaciale, Department of Oral and Maxillofacial Surgery, U1008 - Controlled Drug Delivery Systems and Biomaterials, Université de Lille/CHU de Lille, Lille, France
| | - Joel Ferri
- Professor and Department Head, Stomatologie, Chirurgie Orale et Maxillofaciale, Department of Oral and Maxillofacial Surgery, U1008 - Controlled Drug Delivery Systems and Biomaterials, Université de Lille/CHU de Lille, Lille, France
| | - Constantinus Politis
- Professor and Department Head, Oral and Maxillofacial Surgery - Imaging and Pathology Research Group, Department of Imaging and Pathology, Faculty of Medicine, University of Leuven and Department of Oral & Maxillofacial Surgery, University of Leuven, Leuven, Belgium
| |
Collapse
|
31
|
Yahara Y, Ma X, Gracia L, Alman BA. Monocyte/Macrophage Lineage Cells From Fetal Erythromyeloid Progenitors Orchestrate Bone Remodeling and Repair. Front Cell Dev Biol 2021; 9:622035. [PMID: 33614650 PMCID: PMC7889961 DOI: 10.3389/fcell.2021.622035] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/12/2021] [Indexed: 12/21/2022] Open
Abstract
A third of the population sustains a bone fracture, and the pace of fracture healing slows with age. The slower pace of repair is responsible for the increased morbidity in older individuals who sustain a fracture. Bone healing progresses through overlapping phases, initiated by cells of the monocyte/macrophage lineage. The repair process ends with remodeling. This last phase is controlled by osteoclasts, which are bone-specific multinucleated cells also of the monocyte/macrophage lineage. The slower rate of healing in aging can be rejuvenated by macrophages from young animals, and secreted proteins from macrophage regulate undifferentiated mesenchymal cells to become bone-forming osteoblasts. Macrophages can derive from fetal erythromyeloid progenitors or from adult hematopoietic progenitors. Recent studies show that fetal erythromyeloid progenitors are responsible for the osteoclasts that form the space in bone for hematopoiesis and the fetal osteoclast precursors reside in the spleen postnatally, traveling through the blood to participate in fracture repair. Differences in secreted proteins between macrophages from old and young animals regulate the efficiency of osteoblast differentiation from undifferentiated mesenchymal precursor cells. Interestingly, during the remodeling phase osteoclasts can form from the fusion between monocyte/macrophage lineage cells from the fetal and postnatal precursor populations. Data from single cell RNA sequencing identifies specific markers for populations derived from the different precursor populations, a finding that can be used in future studies. Here, we review the diversity of macrophages and osteoclasts, and discuss recent finding about their developmental origin and functions, which provides novel insights into their roles in bone homeostasis and repair.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, Japan.,Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Xinyi Ma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Liam Gracia
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
32
|
Moore ER, Mathews OA, Yao Y, Yang Y. Prx1-expressing cells contributing to fracture repair require primary cilia for complete healing in mice. Bone 2021; 143:115738. [PMID: 33188955 PMCID: PMC7769995 DOI: 10.1016/j.bone.2020.115738] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/25/2020] [Accepted: 11/07/2020] [Indexed: 02/09/2023]
Abstract
Bone is a dynamic organ that is continuously modified during development, load-induced adaptation, and fracture repair. Understanding the cellular and molecular mechanisms for natural fracture healing can lead to therapeutics that enhance the quality of newly formed tissue, advance the rate of healing, or replace the need for invasive surgical procedures. Prx1-expressing cells in the periosteum are thought to supply the majority of osteoblasts and chondrocytes in the fracture callus, but the exact mechanisms for this behavior are unknown. The primary cilium is a sensory organelle that is known to mediate several signaling pathways involved in fracture healing and required for Prx1-expressing cells to contribute to juvenile bone development and adult load-induced bone formation. We therefore investigated the role of Prx1-expressing cell primary cilia in fracture repair by developing a mouse model that enabled us to simultaneously track Prx1 lineage cell fate and disrupt Prx1-expressing cell primary cilia in vivo. The cilium KO mice exhibited abnormally large calluses with significantly decreased bone formation and persistent cartilage nodules. Analysis of mRNA expression in the early soft callus revealed downregulation of osteogenesis, Hh signaling, and Wnt signaling, and upregulation of chondrogenesis and angiogenesis. The mutant mice also exhibited decreased Osx and Periostin but increased αSMA and PECAM-1 protein expression in the hard callus. We further used a Gli1LacZ reporter and found that Hh signaling was significantly upregulated in the mutant callus at later stages of healing. Interestingly, altered protein expression and Hh signaling did not correlate with labeled Prx1-lineage cells, suggesting loss of cilia altered Hh signaling non-autonomously. Overall, cilium KO mice demonstrated severely delayed and incomplete fracture healing, and our findings suggest Prx1-expressing cell primary cilia are necessary to tune Hh signaling for proper fracture repair.
Collapse
Affiliation(s)
| | - O Amandhi Mathews
- Harvard School of Dental Medicine, Boston, MA, USA; University of Dallas, Irving, TX, USA
| | - Yichen Yao
- Harvard School of Dental Medicine, Boston, MA, USA; Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingzi Yang
- Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
33
|
Rodríguez-Merchán EC. A Review of Recent Developments in the Molecular Mechanisms of Bone Healing. Int J Mol Sci 2021; 22:767. [PMID: 33466612 PMCID: PMC7828700 DOI: 10.3390/ijms22020767] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Between 5 and 10 percent of fractures do not heal, a condition known as nonunion. In clinical practice, stable fracture fixation associated with autologous iliac crest bone graft placement is the gold standard for treatment. However, some recalcitrant nonunions do not resolve satisfactorily with this technique. For these cases, biological alternatives are sought based on the molecular mechanisms of bone healing, whose most recent findings are reviewed in this article. The pro-osteogenic efficacy of morin (a pale yellow crystalline flavonoid pigment found in old fustic and osage orange trees) has recently been reported, and the combined use of bone morphogenetic protein-9 (BMP9) and leptin might improve fracture healing. Inhibition with methyl-piperidino-pyrazole of estrogen receptor alpha signaling delays bone regeneration. Smoking causes a chondrogenic disorder, aberrant activity of the skeleton's stem and progenitor cells, and an intense initial inflammatory response. Smoking cessation 4 weeks before surgery is therefore highly recommended. The delay in fracture consolidation in diabetic animals is related to BMP6 deficiency (35 kDa). The combination of bioceramics and expanded autologous human mesenchymal stem cells from bone marrow is a new and encouraging alternative for treating recalcitrant nonunions.
Collapse
Affiliation(s)
- Emerito Carlos Rodríguez-Merchán
- Osteoarticular Surgery Research, Hospital La Paz Institute for Health Research-IdiPAZ, La Paz University Hospital-Autonomous University of Madrid, 28046 Madrid, Spain
| |
Collapse
|
34
|
Local injections of β-NGF accelerates endochondral fracture repair by promoting cartilage to bone conversion. Sci Rep 2020; 10:22241. [PMID: 33335129 PMCID: PMC7747641 DOI: 10.1038/s41598-020-78983-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
There are currently no pharmacological approaches in fracture healing designed to therapeutically stimulate endochondral ossification. In this study, we test nerve growth factor (NGF) as an understudied therapeutic for fracture repair. We first characterized endogenous expression of Ngf and its receptor tropomyosin receptor kinase A (TrkA) during tibial fracture repair, finding that they peak during the cartilaginous phase. We then tested two injection regimens and found that local β-NGF injections during the endochondral/cartilaginous phase promoted osteogenic marker expression. Gene expression data from β-NGF stimulated cartilage callus explants show a promotion in markers associated with endochondral ossification such as Ihh, Alpl, and Sdf-1. Gene ontology enrichment analysis revealed the promotion of genes associated with Wnt activation, PDGF- and integrin-binding. Subsequent histological analysis confirmed Wnt activation following local β-NGF injections. Finally, we demonstrate functional improvements to bone healing following local β-NGF injections which resulted in a decrease in cartilage and increase of bone volume. Moreover, the newly formed bone contained higher trabecular number, connective density, and bone mineral density. Collectively, we demonstrate β-NGF’s ability to promote endochondral repair in a murine model and uncover mechanisms that will serve to further understand the molecular switches that occur during cartilage to bone transformation.
Collapse
|
35
|
Sun J, Feng H, Xing W, Han Y, Suo J, Yallowitz AR, Qian N, Shi Y, Greenblatt MB, Zou W. Histone demethylase LSD1 is critical for endochondral ossification during bone fracture healing. SCIENCE ADVANCES 2020; 6:6/45/eaaz1410. [PMID: 33148658 PMCID: PMC7673679 DOI: 10.1126/sciadv.aaz1410] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 09/16/2020] [Indexed: 06/11/2023]
Abstract
Bone fracture is repaired predominantly through endochondral ossification. However, the regulation of endochondral ossification by key factors during fracture healing remains largely enigmatic. Here, we identify histone modification enzyme LSD1 as a critical factor regulating endochondral ossification during bone regeneration. Loss of LSD1 in Prx1 lineage cells severely impaired bone fracture healing. Mechanistically, LSD1 tightly controls retinoic acid signaling through regulation of Aldh1a2 expression level. The increased retinoic acid signaling in LSD1-deficient mice suppressed SOX9 expression and impeded the cartilaginous callus formation during fracture repair. The discovery that LSD1 can regulate endochondral ossification during fracture healing will benefit the understanding of bone regeneration and have implications for regenerative medicine.
Collapse
Affiliation(s)
- Jun Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Heng Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Wenhui Xing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yujiao Han
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Jinlong Suo
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Niandong Qian
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Yujiang Shi
- Newborn Medicine Division, Boston Children's Hospital and Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| |
Collapse
|
36
|
Esposito A, Wang L, Li T, Miranda M, Spagnoli A. Role of Prx1-expressing skeletal cells and Prx1-expression in fracture repair. Bone 2020; 139:115521. [PMID: 32629173 PMCID: PMC7484205 DOI: 10.1016/j.bone.2020.115521] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
The healing capacity of bones after fracture implies the existence of adult regenerative cells. However, information on identification and functional role of fracture-induced progenitors is still lacking. Paired-related homeobox 1 (Prx1) is expressed during skeletogenesis. We hypothesize that fracture recapitulates Prx1's expression, and Prx1 expressing cells are critical to induce repair. To address our hypothesis, we used a combination of in vivo and in vitro approaches, short and long-term cell tracking analyses of progenies and actively expressing cells, cell ablation studies, and rodent animal models for normal and defective fracture healing. We found that fracture elicits a periosteal and endosteal response of perivascular Prx1+ cells that participate in fracture healing and showed that Prx1-expressing cells have a functional role in the repair process. While Prx1-derived cells contribute to the callus, Prx1's expression decreases concurrently with differentiation into cartilaginous and bone cells, similarly to when Prx1+ cells are cultured in differentiating conditions. We determined that bone morphogenic protein 2 (BMP2), through C-X-C motif-ligand-12 (CXCL12) signaling, modulates the downregulation of Prx1. We demonstrated that fracture elicits an early increase in BMP2 expression, followed by a decrease in CXCL12 that in turn down-regulates Prx1, allowing cells to commit to osteochondrogenesis. In vivo and in vitro treatment with CXCR4 antagonist AMD3100 restored Prx1 expression by modulating the BMP2-CXCL12 axis. Our studies represent a shift in the current research that has primarily focused on the identification of markers for postnatal skeletal progenitors, and instead we characterized the function of a specific population (Prx1+ cells) and their expression marker (Prx1) as a crossroad in fracture repair. The identification of fracture-induced perivascular Prx1+ cells and regulation of Prx1's expression by BMP2 and in turn by CXCL12 in the orchestration of fracture repair, highlights a pathway in which to investigate defective mechanisms and therapeutic targets for fracture non-union.
Collapse
Affiliation(s)
- Alessandra Esposito
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Tieshi Li
- Department of Pediatrics, University of Nebraska Medical Center, Children's Hospital & Medical Center, Omaha, NE, USA
| | - Mariana Miranda
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Anna Spagnoli
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA; Department of Pediatrics, Division of Pediatric Endocrinology, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
37
|
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
|
38
|
Shoji S, Uchida K, Saito W, Sekiguchi H, Inoue G, Miyagi M, Kuroda A, Takaso M. Acceleration of Bone Healing by In Situ-Forming Dextran-Tyramine Conjugates Containing Basic Fibroblast Growth Factor in Mice. Cureus 2020; 12:e10085. [PMID: 32874816 PMCID: PMC7455394 DOI: 10.7759/cureus.10085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
An enzymatic crosslinking strategy using hydrogen peroxide (H2O2) and horseradish peroxidase (HRP) has been receiving increasing attention for use with in situ-formed hydrogels (IFHs). Several studies have reported the application of IFHs in cell delivery and tissue engineering. IFHs may also be ideal carrier materials for bone repair, although their potential as a carrier for basic fibroblast growth factor (bFGF) has yet to be evaluated. Here, we examined the effect of an IFH made of dextran (Dex)-tyramine (TA) conjugates (IFH-Dex-TA) containing bFGF in promoting bone formation in a fracture model in mice. Immediately following a fracture procedure, animals either received no treatment (control) or an injection of IFH-Dex-TA/phosphate-buffered saline (IFH-Dex-TA/PBS) or IFH-Dex-TA containing 1 μg bFGF (IFH-Dex-TA/bFGF) into the fracture site (n=10, each treatment). Fracture sites injected with IFH-Dex-TA/bFGF showed significantly greater bone volume, mineral content, and bone union than sites receiving no treatment or treated with IFH-Dex-TA/PBS alone (each n=10). This Dex-TA gel may be an effective drug delivery system for optimizing bFGF therapy.
Collapse
Affiliation(s)
- Shintaro Shoji
- Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, JPN
| | - Kentaro Uchida
- Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, JPN
| | - Wataru Saito
- Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, JPN
| | | | - Gen Inoue
- Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, JPN
| | - Masayuki Miyagi
- Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, JPN
| | - Akiyoshi Kuroda
- Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, JPN
| | - Masashi Takaso
- Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, JPN
| |
Collapse
|
39
|
Thorup AS, Dell'Accio F, Eldridge SE. Lessons from joint development for cartilage repair in the clinic. Dev Dyn 2020; 250:360-376. [PMID: 32738003 DOI: 10.1002/dvdy.228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.
Collapse
Affiliation(s)
- Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
40
|
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
|
41
|
Lukač N, Katavić V, Novak S, Šućur A, Filipović M, Kalajzić I, Grčević D, Kovačić N. What do we know about bone morphogenetic proteins and osteochondroprogenitors in inflammatory conditions? Bone 2020; 137:115403. [PMID: 32371019 DOI: 10.1016/j.bone.2020.115403] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Osteochondroprogenitors are crucial for embryonic bone development and postnatal processes such as bone repair in response to fracture injury, and their dysfunction may contribute to insufficient repair of structural damage in inflammatory arthritides. In the fracture healing, the early inflammatory phase is crucial for normal callus development and new bone formation. This process involves a complex interplay of many molecules and cell types, responsible for recruitment, expansion and differentiation of osteochondroprogenitor populations. In inflammatory arthritides, inflammation induces bone resorption and causes insufficient bone formation, which leads to local and systemic bone loss. While bone loss is a predominant feature in rheumatoid arthritis, inflammation also induces pathologic bone formation at enthesial sites in seronegative spondyloarthropathies. Bone morphogenetic proteins (BMP) are involved in cell proliferation, differentiation and apoptosis, and have fundamental roles in maintenance of postnatal bone homeostasis. They are crucial regulators of the osteochondroprogenitor pool and drive their proliferation, differentiation, and lifespan during bone regeneration. In this review, we summarize the effects of inflammation on osteochondroprogenitor populations during fracture repair and in inflammatory arthritides, with special focus on inflammation-mediated modulation of BMP signaling. We also present data in which we describe a population of murine synovial osteochondroprogenitor cells, which are reduced in arthritis, and characterize their expression of genes involved in regulation of bone homeostasis, emphasizing the up-regulation of BMP pathways in early progenitor subset. Based on the presented data, it may be concluded that during an inflammatory response, innate immune cells induce osteochondroprogenitors by providing signals for their recruitment, by producing BMPs and other osteogenic factors for paracrine effects, and by secreting inflammatory cytokines that may positively regulate osteogenic pathways. On the other hand, inflammatory cells may secrete cytokines that interfere with osteogenic pathways, proapoptotic factors that reduce the pool of osteochondroprogenitor cells, as well as BMP and Wnt antagonists. The net effect is strongly context-dependent and influenced by the local milieu of cells, cytokines, and growth factors. Further elucidation of the interplay between inflammatory signals and BMP-mediated bone formation may provide valuable tools for therapeutic targeting.
Collapse
Affiliation(s)
- Nina Lukač
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vedran Katavić
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sanja Novak
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Alan Šućur
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Maša Filipović
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Danka Grčević
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia.
| |
Collapse
|
42
|
Xu GP, Zhang XF, Sun L, Chen EM. Current and future uses of skeletal stem cells for bone regeneration. World J Stem Cells 2020; 12:339-350. [PMID: 32547682 PMCID: PMC7280866 DOI: 10.4252/wjsc.v12.i5.339] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/07/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
The postnatal skeleton undergoes growth, modeling, and remodeling. The human skeleton is a composite of diverse tissue types, including bone, cartilage, fat, fibroblasts, nerves, blood vessels, and hematopoietic cells. Fracture nonunion and bone defects are among the most challenging clinical problems in orthopedic trauma. The incidence of nonunion or bone defects following fractures is increasing. Stem and progenitor cells mediate homeostasis and regeneration in postnatal tissue, including bone tissue. As multipotent stem cells, skeletal stem cells (SSCs) have a strong effect on the growth, differentiation, and repair of bone regeneration. In recent years, a number of important studies have characterized the hierarchy, differential potential, and bone formation of SSCs. Here, we describe studies on and applications of SSCs and/or mesenchymal stem cells for bone regeneration.
Collapse
Affiliation(s)
- Guo-Ping Xu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xiang-Feng Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Lu Sun
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, United States
| | - Er-Man Chen
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
43
|
Touri M, Moztarzadeh F, Abu Osman NA, Dehghan MM, Brouki Milan P, Farzad-Mohajeri S, Mozafari M. Oxygen-Releasing Scaffolds for Accelerated Bone Regeneration. ACS Biomater Sci Eng 2020; 6:2985-2994. [PMID: 33463293 DOI: 10.1021/acsbiomaterials.9b01789] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Hypoxia, the result of disrupted vasculature, can be categorized in the main limiting factors for fracture healing. A lack of oxygen can cause cell apoptosis, tissue necrosis, and late tissue healing. Remedying hypoxia by supplying additional oxygen will majorly accelerate bone healing. In this study, biphasic calcium phosphate (BCP) scaffolds were fabricated by robocasting, an additive manufacturing technique. Then, calcium peroxide (CPO) particles, as an oxygen-releasing agent, were coated on the BCP scaffolds. Segmental radial defects with the size of 15 mm were created in rabbits. Uncoated and CPO-coated BCP scaffolds were implanted in the defects. The empty (control) group received no implantation. Repairing of the bone was investigated via X-ray, histological analysis, and biomechanical tests at 3 and 6 months postoperatively, with immunohistochemical examinations at 6 months after operation. According to the radiological observations, formation of new bone was augmented at the interface between the implant and host bone and internal pores of CPO-coated BCP scaffolds compared to uncoated scaffolds. Histomorphometry analysis represented that the amount of newly formed bone in the CPO-coated scaffold was nearly two times higher than the uncoated one. Immunofluorescence staining revealed that osteogenic markers, osteonectin and octeocalcin, were overexpressed in the defects treated with the coated scaffolds at 6 months of postsurgery, demonstrating higher osteogenic differentiation and bone mineralization compared to the uncoated scaffold group. Furthermore, the coated scaffolds had superior biomechanical properties as in the case of 3 months after surgery, the maximal flexural force of the coated scaffolds reached to 134 N, while it was 92 N for uncoated scaffolds. The results could assure a boosted ability of bone repair for CPO-coated BCP scaffolds implanted in the segmental defect of rabbit radius because of oxygen-releasing coating, and this system of oxygen-generating coating/scaffold might be a potential for accelerated repairing of bone defects.
Collapse
Affiliation(s)
- Maria Touri
- Biomaterial Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran 1591634311, Iran
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Fathollah Moztarzadeh
- Biomaterial Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran 1591634311, Iran
| | - Noor Azuan Abu Osman
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417466191, Iran
- Institute of Biomedical Research, University of Tehran, Tehran 1417466191, Iran
| | - Peiman Brouki Milan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | | | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| |
Collapse
|
44
|
Serowoky MA, Arata CE, Crump JG, Mariani FV. Skeletal stem cells: insights into maintaining and regenerating the skeleton. Development 2020; 147:147/5/dev179325. [PMID: 32161063 DOI: 10.1242/dev.179325] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Skeletal stem cells (SSCs) generate the progenitors needed for growth, maintenance and repair of the skeleton. Historically, SSCs have been defined as bone marrow-derived cells with inconsistent characteristics. However, recent in vivo tracking experiments have revealed the presence of SSCs not only within the bone marrow but also within the periosteum and growth plate reserve zone. These studies show that SSCs are highly heterogeneous with regard to lineage potential. It has also been revealed that, during digit tip regeneration and in some non-mammalian vertebrates, the dedifferentiation of osteoblasts may contribute to skeletal regeneration. Here, we examine how these research findings have furthered our understanding of the diversity and plasticity of SSCs that mediate skeletal maintenance and repair.
Collapse
Affiliation(s)
- Maxwell A Serowoky
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Claire E Arata
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Francesca V Mariani
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
45
|
Yokoi H, Take Y, Uchida R, Magome T, Shimomura K, Mae T, Okamoto T, Hanai T, Chong Y, Sato S, Hikida M, Nakata K. Vibration acceleration promotes endochondral formation during fracture healing through cellular chondrogenic differentiation. PLoS One 2020; 15:e0229127. [PMID: 32134943 PMCID: PMC7058294 DOI: 10.1371/journal.pone.0229127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
Vibration acceleration through whole body vibration has been reported to promote fracture healing. However, the mechanism responsible for this effect remains unclear. Purpose of this study was to determine whether vibration acceleration directly affects cells around the fracture site and promotes endochondral ossification. Four-week-old female Wistar Hannover rats were divided into two groups (vibration [V group] and control [C group]). The eighth ribs on both sides were cut vertically using scissors. From postoperative day 3 to 11, vibration acceleration using Power Plate® (30 Hz, low amplitude [30-Low], 10 min/day) was applied in the V group. Mature calluses appeared earlier in the V group than in the C group by histological analysis. The GAG content in the fracture callus on day 6 was significantly higher in the V group than in the C group. The mRNA expressions of SOX-9, aggrecan, and Col-II in the fracture callus on day 6 and Col-X on day 9 were significantly higher in the V group than in the C group. For in vitro analysis, four different conditions of vibration acceleration (30 or 50 Hz with low or high amplitude [30-Low, 30-High, 50-Low, and 50-High], 10 min/day) were applied to a prechondrogenic cell (ATDC5) and an undifferentiated cell (C3H10T1/2). There was no significant difference in cell proliferation between the control and any of the four vibration conditions for both cell lines. For both cell lines, alcian blue staining was greater under 30-Low and 50-Low conditions than under control as well as 30-High and 50-High conditions on days 7 and 14. Vibration acceleration under 30-L condition upregulated chondrogenic gene expressions of SOX-9, aggrecan, Col-II, and Col-X. Low-amplitude vibration acceleration can promote endochondral ossification in the fracture healing in vivo and chondrogenic differentiation in vitro.
Collapse
Affiliation(s)
- Hiroyuki Yokoi
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasuhiro Take
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryohei Uchida
- Department of Sports Medicine, Yukioka Hospital, Osaka, Japan
| | - Takuya Magome
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazunori Shimomura
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tatsuo Mae
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomoko Okamoto
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tatsuhiro Hanai
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yang Chong
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Seira Sato
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Minami Hikida
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ken Nakata
- Medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
- * E-mail:
| |
Collapse
|
46
|
Ying J, Xu T, Wang C, Jin H, Tong P, Guan J, Abu-Amer Y, O’Keefe R, Shen J. Dnmt3b ablation impairs fracture repair through upregulation of Notch pathway. JCI Insight 2020; 5:131816. [PMID: 32051335 PMCID: PMC7098799 DOI: 10.1172/jci.insight.131816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
We previously established that DNA methyltransferase 3b (Dnmt3b) is the sole Dnmt responsive to fracture repair and that Dnmt3b expression is induced in progenitor cells during fracture repair. In the current study, we confirmed that Dnmt3b ablation in mesenchymal progenitor cells (MPCs) resulted in impaired endochondral ossification, delayed fracture repair, and reduced mechanical strength of the newly formed bone in Prx1-Cre;Dnmt3bf/f (Dnmt3bPrx1) mice. Mechanistically, deletion of Dnmt3b in MPCs led to reduced chondrogenic and osteogenic differentiation in vitro. We further identified Rbpjκ as a downstream target of Dnmt3b in MPCs. In fact, we located 2 Dnmt3b binding sites in the murine proximal Rbpjκ promoter and gene body and confirmed Dnmt3b interaction with the 2 binding sites by ChIP assays. Luciferase assays showed functional utilization of the Dnmt3b binding sites in murine C3H10T1/2 cells. Importantly, we showed that the MPC differentiation defect observed in Dnmt3b deficiency cells was due to the upregulation of Rbpjκ, evident by restored MPC differentiation upon Rbpjκ inhibition. Consistent with in vitro findings, Rbpjκ blockage via dual antiplatelet therapy reversed the differentiation defect and accelerated fracture repair in Dnmt3bPrx1 mice. Collectively, our data suggest that Dnmt3b suppresses Notch signaling during MPC differentiation and is necessary for normal fracture repair.
Collapse
Affiliation(s)
- Jun Ying
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Taotao Xu
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuicui Wang
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianjun Guan
- Department of Biomedical Engineering, School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Shriners Hospital for Children, St. Louis, Missouri, USA
| | - Regis O’Keefe
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
47
|
Bone Regeneration, Reconstruction and Use of Osteogenic Cells; from Basic Knowledge, Animal Models to Clinical Trials. J Clin Med 2020; 9:jcm9010139. [PMID: 31947922 PMCID: PMC7019836 DOI: 10.3390/jcm9010139] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/21/2019] [Accepted: 01/02/2020] [Indexed: 01/01/2023] Open
Abstract
The deterioration of the human skeleton's capacity for self-renewal occurs naturally with age. Osteoporosis affects millions worldwide, with current treatments including pharmaceutical agents that target bone formation and/or resorption. Nevertheless, these clinical approaches often result in long-term side effects, with better alternatives being constantly researched. Mesenchymal stem cells (MSCs) derived from bone marrow and adipose tissue are known to hold therapeutic value for the treatment of a variety of bone diseases. The following review summarizes the latest studies and clinical trials related to the use of MSCs, both individually and combined with other methods, in the treatment of a variety of conditions related to skeletal health. For example, some of the most recent works noted the advantage of bone grafts based on biomimetic scaffolds combined with MSC and growth factor delivery, with a greatly increased regeneration rate and minimized side effects for patients. This review also highlights the continuing research into the mechanisms underlying bone homeostasis, including the key transcription factors and signalling pathways responsible for regulating the differentiation of osteoblast lineage. Paracrine factors and specific miRNAs are also believed to play a part in MSC differentiation. Furthering the understanding of the specific mechanisms of cellular signalling in skeletal remodelling is key to incorporating new and effective treatment methods for bone disease.
Collapse
|
48
|
Wang D, Gilbert JR, Zhang X, Zhao B, Ker DFE, Cooper GM. Calvarial Versus Long Bone: Implications for Tailoring Skeletal Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2019; 26:46-63. [PMID: 31588853 DOI: 10.1089/ten.teb.2018.0353] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue-engineered graft substitutes have shown great potential to treat large bone defects. While we usually assume that therapeutic approaches developed for appendicular bone healing could be similarly translated for application in craniofacial reconstruction and vice versa, this is not necessarily accurate. In addition to those more well-known healing-associated factors, such as age, lifestyle (e.g., nutrition and smoking), preexisting disease (e.g., diabetes), medication, and poor blood supply, the developmental origins and surrounding tissue of the wound sites can largely affect the fracture healing outcome as well as designed treatments. Therefore, the strategies developed for long bone fracture repair might not be suitable or directly applicable to skull bone repair. In this review, we discuss aspects of development, healing process, structure, and tissue engineering considerations between calvarial and long bones to assist in designing the tailored bone repair strategies. Impact Statement We summarized, in this review, the existing body of knowledge between long bone and calvarial bone with regard to their development and healing, tissue structure, and consideration of current tissue engineering strategies. By highlighting their similarities and differences, we propose that tailored tissue engineering strategies, such as scaffold features, growth factor usage, and the source of cells for tissue or region-specific bone repair, are necessary to ensure an optimized healing outcome.
Collapse
Affiliation(s)
- Dan Wang
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James R Gilbert
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingkun Zhao
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gregory M Cooper
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
49
|
Doherty L, Yu J, Wang X, Hankenson KD, Kalajzic I, Sanjay A. A PDGFRβ-PI3K signaling axis mediates periosteal cell activation during fracture healing. PLoS One 2019; 14:e0223846. [PMID: 31665177 PMCID: PMC6821073 DOI: 10.1371/journal.pone.0223846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022] Open
Abstract
Insufficient and delayed fracture healing remain significant public health problems with limited therapeutic options. Phosphoinositide 3-kinase (PI3K) signaling, a major pathway involved in regulation of fracture healing, promotes proliferation, migration, and differentiation of osteoprogenitors. We have recently reported that knock-in mice with a global increase in PI3K signaling (gCblYF) show enhanced femoral fracture healing characterized by an extraordinary periosteal response to injury. Interestingly, of all growth factor receptors involved in fracture healing, PI3K directly binds only to PDGFR. Given these findings, we hypothesized a PDGFR-PI3K interaction is necessary for mediating robust periosteal cell activation following fracture. In this study, we isolated primary periosteal cells from gCblYF mice to analyze cross-talk between the PDGFRβ and PI3K signaling pathways. We found PDGFRβ signaling contributes to robust Akt phosphorylation in periosteal cells in comparison with other growth factor signaling pathways. Additionally, we performed femoral fractures on gCblYF mice with a conditional removal of PDGFRβ in mesenchymal progenitors using inducible alpha smooth muscle actin (αSMA) CreERT2 mice. Our studies showed that depletion of PDGFRβ signaling within these progenitors in the early phase of fracture healing significantly abrogates PI3K-mediated periosteal activation and proliferation three days after fracture. Combined, these results suggest that PDGFRβ signaling through PI3K is necessary for robust periosteal activation in the earliest phases of fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
| | - Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
| | - Xi Wang
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, United States of America
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, United States of America
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
50
|
Differential fracture response to traumatic brain injury suggests dominance of neuroinflammatory response in polytrauma. Sci Rep 2019; 9:12199. [PMID: 31434912 PMCID: PMC6704103 DOI: 10.1038/s41598-019-48126-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/24/2019] [Indexed: 12/16/2022] Open
Abstract
Polytraumatic injuries, specifically long bone fracture and traumatic brain injury (TBI), frequently occur together. Clinical observation has long held that TBI can accelerate fracture healing, yet the complexity and heterogeneity of these injuries has produced conflicting data with limited information on underlying mechanisms. We developed a murine polytrauma model with TBI and fracture to evaluate healing in a controlled system. Fractures were created both contralateral and ipsilateral to the TBI to test whether differential responses of humoral and/or neuronal systems drove altered healing patterns. Our results show increased bone formation after TBI when injuries occur contralateral to each other, rather than ipsilateral, suggesting a role of the nervous system based on the crossed neuroanatomy of motor and sensory systems. Analysis of the humoral system shows that blood cell counts and inflammatory markers are differentially modulated by polytrauma. A data-driven multivariate analysis integrating all outcome measures showed a distinct pathological state of polytrauma and co-variations between fracture, TBI and systemic markers. Taken together, our results suggest that a contralateral bone fracture and TBI alter the local neuroinflammatory state to accelerate early fracture healing. We believe applying a similar data-driven approach to clinical polytrauma may help to better understand the complicated pathophysiological mechanisms of healing.
Collapse
|