1
|
Wang X, Wang S, Mu H, Yang C, Dong W, Wang X, Wang J. Macrophage-derived amphiregulin promoted the osteogenic differentiation of chondrocytes through EGFR/Yap axis and TGF-β activation. Bone 2024; 190:117275. [PMID: 39383984 DOI: 10.1016/j.bone.2024.117275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/15/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Endochondral ossification represents a crucial biological process in skeletal development and bone defect repair. Macrophages, recognized as key players in the immune system, are now acknowledged for their substantial role in promoting endochondral ossification within cartilage. Concurrently, the epidermal growth factor receptor (EGFR) ligand amphiregulin (Areg) has been documented for its contributory role in restoring bone tissue homeostasis post-injury. However, the mechanism by which macrophage-secreted Areg facilitates bone repair remains elusive. In this study, the induction of macrophage depletion through in vivo administration of clodronate liposomes was employed in a standard open tibial fracture mouse model to assess bone healing using micro-computed tomography (micro-CT) analysis, histomorphology, and ELISA serum evaluations. The investigation revealed sustained expression of Areg during the fracture healing period in wild-type mice. Macrophage depletion significantly reduced the number of macrophages on the local bone surface and vital organs. This reduction led to diminished Areg secretion, decreased collagen production, and delayed fracture healing. However, histological and micro-CT assessments at 7 and 21 days post-local Areg treatment exhibited a marked improvement of bone healing compared to the vehicle control. In vitro studies demonstrated an increase of Areg secretion by the Raw264.7 cells upon ATP stimulation. Indirect co-culture of Raw264.7 and ATDC5 cells indicated that Areg overexpression enhanced the osteogenic potential of chondrocytes, and vice versa. This osteogenic promotion was attributed to Areg's activation of the membrane receptor EGFR in the ATDC5 cell line, the enhanced phosphorylation of transcription factor Yap, and the facilitation of the expression of bioactive TGF-β by chondrocytes. Collectively, this research elucidates the direct mechanistic effects of macrophage-secreted Areg in promoting bone homeostasis following bone injury.
Collapse
Affiliation(s)
- Xinyi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Shuo Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Hailin Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chang Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Xinru Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jaiwei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|
2
|
Wu W, Zhao Z, Wang Y, Liu M, Zhu G, Li L. Mechanism research of elastic fixation promoting fracture healing based on proteomics and fracture microenvironment. Bone Joint Res 2024; 13:559-572. [PMID: 39377775 PMCID: PMC11460404 DOI: 10.1302/2046-3758.1310.bjr-2023-0257.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/09/2024] Open
Abstract
Aims This study aimed to demonstrate the promoting effect of elastic fixation on fracture, and further explore its mechanism at the gene and protein expression levels. Methods A closed tibial fracture model was established using 12 male Japanese white rabbits, and divided into elastic and stiff fixation groups based on different fixation methods. Two weeks after the operation, a radiograph and pathological examination of callus tissue were used to evaluate fracture healing. Then, the differentially expressed proteins (DEPs) were examined in the callus using proteomics. Finally, in vitro cell experiments were conducted to investigate hub proteins involved in this process. Results Mean callus volume was larger in the elastic fixation group (1,755 mm3 (standard error of the mean (SEM) 297)) than in the stiff fixation group (258 mm3 (SEM 65)). Pathological observation found that the expression levels of osterix (OSX), collagen, type I, alpha 1 (COL1α1), and alkaline phosphatase (ALP) in the callus of the elastic fixation group were higher than those of the stiff fixation group. The protein sequence of the callus revealed 199 DEPs, 124 of which were highly expressed in the elastic fixation group. In the in vitro study, it was observed that a stress of 200 g led to upregulation of thrombospondin 1 (THBS1) and osteoglycin (OGN) expression in bone marrow mesenchymal stem cells (BMSCs). Additionally, these genes were found to be upregulated during the osteogenic differentiation process of the BMSCs. Conclusion Elastic fixation can promote fracture healing and osteoblast differentiation in callus, and the ability of elastic fixation to promote osteogenic differentiation of BMSCs may be achieved by upregulating genes such as THBS1 and OGN.
Collapse
Affiliation(s)
- Weiyong Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihui Zhao
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yongqing Wang
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Meiyue Liu
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Genbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
3
|
Romanowicz GE, Zhang L, Bolger MW, Lynch M, Kohn DH. Beyond bone volume: Understanding tissue-level quality in healing of maxillary vs. femoral defects. Acta Biomater 2024; 187:409-421. [PMID: 39214162 DOI: 10.1016/j.actbio.2024.08.042] [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: 04/04/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Currently, principles of tissue engineering and implantology are uniformly applied to all bone sites, disregarding inherent differences in collagen, mineral composition, and healing rates between craniofacial and long bones. These differences could potentially influence bone quality during the healing process. Evaluating bone quality during healing is crucial for understanding local mechanical properties in regeneration and implant osseointegration. However, site-specific changes in bone quality during healing remain poorly understood. In this study, we assessed newly formed bone quality in sub-critical defects in the maxilla and femur, while impairing collagen cross-linking using β-aminopropionitrile (BAPN). Our findings revealed that femoral healing bone exhibited a 73 % increase in bone volume but showed significantly greater viscoelastic and collagen changes compared to surrounding bone, leading to increased deformation during long-term loading and poorer bone quality in early healing. In contrast, the healing maxilla maintained equivalent hardness and viscoelastic constants compared to surrounding bone, with minimal new bone formation and consistent bone quality. However, BAPN-impaired collagen cross-linking induced viscoelastic changes in the healing maxilla, with no further changes observed in the femur. These results challenge the conventional belief that increased bone volume correlates with enhanced tissue-level bone quality, providing crucial insights for tissue engineering and site-specific implant strategies. The observed differences in bone quality between sites underscore the need for a nuanced approach in assessing the success of regeneration and implant designs and emphasize the importance of exploring site-specific tissue engineering interventions. STATEMENT OF SIGNIFICANCE: Accurate measurement of bone quality is crucial for tissue engineering and implant therapies. Bone quality varies between craniofacial and long bones, yet it's often overlooked in the healing process. Our study is the first to comprehensively analyze bone quality during healing in both the maxilla and femur. Surprisingly, despite significant volume increase, femur healing bone had poorer quality compared to the surrounding bone. Conversely, maxilla healing bone maintained consistent quality despite minimal bone formation. Impaired collagen diminished maxillary healing bone quality, but had no further effect on femur bone quality. These findings challenge the notion that more bone volume equals better quality, offering insights for improving tissue engineering and implant strategies for different bone sites.
Collapse
Affiliation(s)
- Genevieve E Romanowicz
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Lizhong Zhang
- Department of Biomedical Engineering, College of Engineering, University of Michigan, MI, USA
| | - Morgan W Bolger
- Department of Biomedical Engineering, College of Engineering, University of Michigan, MI, USA
| | - Michelle Lynch
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - David H Kohn
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, MI, USA.
| |
Collapse
|
4
|
López JM. Bone Development and Growth. Int J Mol Sci 2024; 25:6767. [PMID: 38928471 PMCID: PMC11203555 DOI: 10.3390/ijms25126767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Our skeleton is an essential part of our body consisting of 206 pieces made of a specialized form of connective tissue, with a matrix containing collagen fibers and a large amount of minerals [...].
Collapse
Affiliation(s)
- José M López
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
5
|
Chen H, Ding Y, Wang Y, Sun Y. The Contribution of Meckel's Cartilage-Derived Type II Collagen-Positive Cells to the Jawbone Development and Repair. J Histochem Cytochem 2024; 72:221554241259059. [PMID: 38836522 PMCID: PMC11179589 DOI: 10.1369/00221554241259059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024] Open
Abstract
Jawbones and long bones, despite their shared skeletal lineage, frequently exhibit distinct origins and developmental pathways. Identifying specific progenitor subsets for mandibular osteogenesis remains challenging. Type II collagen is conventionally associated with cartilaginous structures, yet our investigation has identified the presence of type II collagen positive (Col2+) cells within the jawbone development and regeneration. The role of Col2+ cells in jawbone morphogenesis and repair has remained enigmatic. In this study, we analyze single-cell RNA sequencing data from mice jawbone at embryonic day 10.5. Through fate-mapping experiments, we have elucidated that Col2+ cells and their progeny are instrumental in mandibular osteogenesis across both fetal and postnatal stages. Furthermore, lineage tracing with a tamoxifen-inducible CreER system has established the pivotal role of Col2+ cells, marked by Col2-CreER and originating from the primordial Meckel's cartilage, in jawbone formation. Moreover, our research explored models simulating jawbone defects and tooth extraction, which underscored the osteogenic differentiation capabilities of postnatal Col2+ cells during repair. This finding not only highlights the regenerative potential of Col2+ cells but also suggests their versatility in contributing to skeletal healing and regeneration. In conclusion, our findings position Col2+ cells as essential in orchestrating osteogenesis throughout the continuum of mandibular development and repair.
Collapse
Affiliation(s)
- Hongli Chen
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yunpeng Ding
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yu Wang
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yao Sun
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
6
|
Wu W, Zhao Z, Wang Y, Zhu G, Tan K, Liu M, Li L. Biomechanical Effects of Mechanical Stress on Cells Involved in Fracture Healing. Orthop Surg 2024; 16:811-820. [PMID: 38439564 PMCID: PMC10984830 DOI: 10.1111/os.14026] [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: 10/20/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
Fracture healing is a complex staged repair process in which the mechanical environment plays a key role. Bone tissue is very sensitive to mechanical stress stimuli, and the literature suggests that appropriate stress can promote fracture healing by altering cellular function. However, fracture healing is a coupled process involving multiple cell types that balance and limit each other to ensure proper fracture healing. The main cells that function during different stages of fracture healing are different, and the types and molecular mechanisms of stress required are also different. Most previous studies have used a single mechanical stimulus on individual mechanosensitive cells, and there is no relatively uniform standard for the size and frequency of the mechanical stress. Analyzing the mechanisms underlying the effects of mechanical stimulation on the metabolic regulation of signaling pathways in cells such as in bone marrow mesenchymal stem cells (BMSCs), osteoblasts, chondrocytes, and osteoclasts is currently a challenging research hotspot. Grasping how stress affects the function of different cells at the molecular biology level can contribute to the refined management of fracture healing. Therefore, in this review, we summarize the relevant literature and describe the effects of mechanical stress on cells associated with fracture healing, and their possible signaling pathways, for the treatment of fractures and the further development of regenerative medicine.
Collapse
Affiliation(s)
- Weiyong Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Zhao
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yongqing Wang
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Gengbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Meiyue Liu
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
7
|
Steltzer SS, Abraham AC, Killian ML. Interfacial Tissue Regeneration with Bone. Curr Osteoporos Rep 2024; 22:290-298. [PMID: 38358401 PMCID: PMC11060924 DOI: 10.1007/s11914-024-00859-1] [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] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE OF REVIEW Interfacial tissue exists throughout the body at cartilage-to-bone (osteochondral interface) and tendon-to-bone (enthesis) interfaces. Healing of interfacial tissues is a current challenge in regenerative approaches because the interface plays a critical role in stabilizing and distributing the mechanical stress between soft tissues (e.g., cartilage and tendon) and bone. The purpose of this review is to identify new directions in the field of interfacial tissue development and physiology that can guide future regenerative strategies for improving post-injury healing. RECENT FINDINGS Cues from interfacial tissue development may guide regeneration including biological cues such as cell phenotype and growth factor signaling; structural cues such as extracellular matrix (ECM) deposition, ECM, and cell alignment; and mechanical cues such as compression, tension, shear, and the stiffness of the cellular microenvironment. In this review, we explore new discoveries in the field of interfacial biology related to ECM remodeling, cellular metabolism, and fate. Based on emergent findings across multiple disciplines, we lay out a framework for future innovations in the design of engineered strategies for interface regeneration. Many of the key mechanisms essential for interfacial tissue development and adaptation have high potential for improving outcomes in the clinic.
Collapse
Affiliation(s)
- Stephanie S Steltzer
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Adam C Abraham
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Megan L Killian
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Zhu J, Zhang S, Jin S, Huang C, Shi B, Chen Z, Ji W. Endochondral Repair of Jawbone Defects Using Periosteal Cell Spheroids. J Dent Res 2024; 103:31-41. [PMID: 37968792 DOI: 10.1177/00220345231205273] [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] [Indexed: 11/17/2023] Open
Abstract
Recapitulation of the natural healing process is receiving increasing recognition as a strategy to induce robust tissue regeneration. Endochondral ossification has been recognized as an essential reparative approach in natural jawbone defect healing. However, such an approach has been overlooked in the recent development of cell-based therapeutics for jawbone repair. Therefore, this study aimed to explore a bioinspired stem cell-based strategy for jawbone repair by mimicking the mesenchymal condensation of progenitor cells during the early endochondral ossification process. For this purpose, passage 3 of jawbone periosteum-derived cells (jb-PDCs) was cultured in our previously reported nonadherent microwells (200 µm in diameter, 148 µm in depth, and 100 µm space in between) and self-assembled into spheroids with a diameter of 96.4 ± 5.8 µm after 48 h. Compared to monolayer culture, the jb-PDC spheroids showed a significant reduction of stemness marker expression evidenced by flow cytometry. Furthermore, a significant upregulation of chondrogenic transcription factor SOX9 in both gene and protein levels was observed in the jb-PDC spheroids after 48 h of chondrogenic induction. RNA sequencing and Western blotting analysis further suggested that the enhanced SOX9-mediated chondrogenic differentiation in jb-PDC spheroids was attributed to the activation of the p38 MAPK pathway. Impressively, inhibition of p38 kinase activity significantly attenuated chondrogenic differentiation jb-PDC spheroids, evidenced by a significant decline of SOX9 in both gene and protein levels. Strikingly, the jb-PDC spheroids implanted in 6- to 8-wk-old male C57BL/6 mice with critical-size jawbone defects (1.8 mm in diameter) showed an evident contribution to cartilaginous callus formation after 1 wk, evidenced by histological analysis. Furthermore, micro-computed tomography analysis showed that the jb-PDC spheroids significantly accelerated bone healing after 2 wk in the absence of exogenous growth factors. In sum, the presented findings represent the successful development of cell-based therapeutics to reengineer the endochondral bone repair process and illustrate the potential application to improve bone repair and regeneration in the craniofacial skeleton.
Collapse
Affiliation(s)
- J Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - S Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - S Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - C Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - B Shi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Z Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - W Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
9
|
Carmon I, Zobrab A, Alterman M, Tabib R, Cohen A, Kandel L, Greenberg A, Reich E, Casap N, Dvir-Ginzberg M. Chondrocytes supplemented to bone graft-containing scaffolds expedite cranial defect repair. Sci Rep 2023; 13:19192. [PMID: 37932515 PMCID: PMC10628268 DOI: 10.1038/s41598-023-46604-z] [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: 08/11/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023] Open
Abstract
Critical maxillofacial bone fractures do not heal spontaneously, thus, often there is a need to facilitate repair via surgical intervention. Gold standard approaches, include the use of autologous bone graft, or devices supplemented with osteogenic growth factors and bone substitutes. This research aimed to employ a critical size calvaria defect model, to determine if the addition of chondrocytes to collagen-containing bone graft substitute, may expedite bone repair. As such, using a critical size rat calvaria defect, we implanted a collagen scaffold containing bone graft substitute (i.e., Bone graft scaffold, BG) or BG supplemented with costal chondrocytes (cBG). The rats were subjected to live CT imaging at 1, 6, 9, and 12 weeks following the surgical procedure and sacrificed for microCT imaging of the defect site. Moreover, serum markers and histological evaluation were assessed to determine osseous tissue regeneration and turnover. Live CT and microCT indicated cBG implants displayed expedited bone repair vs, BG alone, already at 6 weeks post defect induction. cBG also displayed a shorter distance between the defect edges and greater mineral apposition distance compared to BG. Summerizing, the data support the addition of chondrocytes to bone substitute, accelerates the formation of new bone within a critical size defect.
Collapse
Affiliation(s)
- Idan Carmon
- Laboratory of Cartilage Biology, Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna Zobrab
- Laboratory of Cartilage Biology, Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael Alterman
- Department of Maxillofacial Surgery, Faculty of Dental Medicine, Hadassah-Hebrew University, P. O. Box 12272, 9112102, Jerusalem, Israel
| | - Rami Tabib
- Department of Maxillofacial Surgery, Faculty of Dental Medicine, Hadassah-Hebrew University, P. O. Box 12272, 9112102, Jerusalem, Israel
| | - Adir Cohen
- Department of Maxillofacial Surgery, Faculty of Dental Medicine, Hadassah-Hebrew University, P. O. Box 12272, 9112102, Jerusalem, Israel
| | - Leonid Kandel
- Joint Replacement and Reconstruction Unit, Orthopedic Surgery Complex, Hadassah-Hebrew University Medical Center at Mount Scopus, Jerusalem, Israel
| | - Alexander Greenberg
- Joint Replacement and Reconstruction Unit, Orthopedic Surgery Complex, Hadassah-Hebrew University Medical Center at Mount Scopus, Jerusalem, Israel
| | - Eli Reich
- Laboratory of Cartilage Biology, Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nardi Casap
- Department of Maxillofacial Surgery, Faculty of Dental Medicine, Hadassah-Hebrew University, P. O. Box 12272, 9112102, Jerusalem, Israel.
| | - Mona Dvir-Ginzberg
- Laboratory of Cartilage Biology, Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
10
|
Smeriglio P, Zalc A. Cranial Neural Crest Cells Contribution to Craniofacial Bone Development and Regeneration. Curr Osteoporos Rep 2023; 21:624-631. [PMID: 37421571 DOI: 10.1007/s11914-023-00804-8] [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] [Accepted: 06/14/2023] [Indexed: 07/10/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarize (i) the latest evidence on cranial neural crest cells (CNCC) contribution to craniofacial development and ossification; (ii) the recent discoveries on the mechanisms responsible for their plasticity; and (iii) the newest procedures to ameliorate maxillofacial tissue repair. RECENT FINDINGS CNCC display a remarkable differentiation potential that exceeds the capacity of their germ layer of origin. The mechanisms by which they expand their plasticity was recently described. Their ability to participate to craniofacial bone development and regeneration open new perspectives for treatments of traumatic craniofacial injuries or congenital syndromes. These conditions can be life-threatening, require invasive maxillofacial surgery and can leave deep sequels on our health or quality of life. With accumulating evidence showing how CNCC-derived stem cells potential can ameliorate craniofacial reconstruction and tissue repair, we believe a deeper understanding of the mechanisms regulating CNCC plasticity is essential to ameliorate endogenous regeneration and improve tissue repair therapies.
Collapse
Affiliation(s)
- Piera Smeriglio
- Centre de Recherche en Myologie, Institut de Myologie, INSERM, Sorbonne Université, 75013, Paris, France.
| | - Antoine Zalc
- Institut Cochin, CNRS, INSERM, Université Paris Cité, 75014, Paris, France.
| |
Collapse
|
11
|
Rundle CH, Gomez GA, Pourteymoor S, Mohan S. Sequential application of small molecule therapy enhances chondrogenesis and angiogenesis in murine segmental defect bone repair. J Orthop Res 2023; 41:1471-1481. [PMID: 36448182 PMCID: PMC10506518 DOI: 10.1002/jor.25493] [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: 04/06/2022] [Revised: 10/03/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
The increasing incidence of physiologic/pathologic conditions that impair the otherwise routine healing of endochondral bone fractures and the occurrence of severe bone injuries necessitate novel approaches to enhance clinically challenging bone fracture repair. To promote the healing of nonunion fractures, we tested an approach that used two small molecules to sequentially enhance cartilage development and conversion to the bone in the callus of a murine femoral segmental defect nonunion model of bone injury. Systemic injections of smoothened agonist 21k (SAG21k) were used to stimulate chondrogenesis through the activation of the sonic hedgehog (SHH) pathway early in bone repair, while injections of the prolyl hydroxylase domain (PHD)2 inhibitor, IOX2, were used to stimulate hypoxia signaling-mediated endochondral bone formation. The expression of SHH pathway genes and Phd2 target genes was increased in chondrocyte cell lines in response to SAG21k and IOX2 treatment, respectively. The segmental defect responded to sequential systemic administration of these small molecules with increased chondrocyte expression of PTCH1, GLI1, and SOX9 in response to SAG and increased expression of hypoxia-induced factor-1α and vascular endothelial growth factor-A in the defect tissues in response to IOX2. At 6 weeks postsurgery, the combined SAG-IOX2 therapy produced increased bone formation in the defect with the bony union over the injury. Clinical significance: This therapeutic approach was successful in promoting cartilage and bone formation within a critical-size segmental defect and established the utility of a sequential small molecule therapy for the enhancement of fracture callus development in clinically challenging bone injuries.
Collapse
Affiliation(s)
- Charles H. Rundle
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
- Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Gustavo A. Gomez
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
- Department of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
12
|
Soares AP, Fischer H, Aydin S, Steffen C, Schmidt-Bleek K, Rendenbach C. Uncovering the unique characteristics of the mandible to improve clinical approaches to mandibular regeneration. Front Physiol 2023; 14:1152301. [PMID: 37008011 PMCID: PMC10063818 DOI: 10.3389/fphys.2023.1152301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
The mandible (lower jaw) bone is aesthetically responsible for shaping the lower face, physiologically in charge of the masticatory movements, and phonetically accountable for the articulation of different phonemes. Thus, pathologies that result in great damage to the mandible severely impact the lives of patients. Mandibular reconstruction techniques are mainly based on the use of flaps, most notably free vascularized fibula flaps. However, the mandible is a craniofacial bone with unique characteristics. Its morphogenesis, morphology, physiology, biomechanics, genetic profile, and osteoimmune environment are different from any other non-craniofacial bone. This fact is especially important to consider during mandibular reconstruction, as all these differences result in unique clinical traits of the mandible that can impact the results of jaw reconstructions. Furthermore, overall changes in the mandible and the flap post-reconstruction may be dissimilar, and the replacement process of the bone graft tissue during healing can take years, which in some cases can result in postsurgical complications. Therefore, the present review highlights the uniqueness of the jaw and how this factor can influence the outcome of its reconstruction while using an exemplary clinical case of pseudoarthrosis in a free vascularized fibula flap.
Collapse
Affiliation(s)
- Ana Prates Soares
- Department of Oral and Maxillofacial Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Ana Prates Soares,
| | - Heilwig Fischer
- Department of Oral and Maxillofacial Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Centrum für Muskuloskeletale Chirurgie, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Sabrin Aydin
- Department of Oral and Maxillofacial Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claudius Steffen
- Department of Oral and Maxillofacial Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Rendenbach
- Department of Oral and Maxillofacial Surgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
13
|
Marcucio R, Miclau T, Bahney C. A Shifting Paradigm: Transformation of Cartilage to Bone during Bone Repair. J Dent Res 2023; 102:13-20. [PMID: 36303415 PMCID: PMC9791286 DOI: 10.1177/00220345221125401] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
While formation and regeneration of the skeleton have been studied for a long period of time, significant scientific advances in this field continue to emerge based on an unmet clinical need to improve options to promote bone repair. In this review, we discuss the relationship between mechanisms of bone formation and bone regeneration. Data clearly show that regeneration is not simply a reinduction of the molecular and cellular programs that were used for development. Instead, the mechanical environment exerts a strong influence on the mode of repair, while during development, cell-intrinsic processes drive the mode of skeletal formation. A major advance in the field has shown that cell fate is flexible, rather than terminal, and that chondrocytes are able to differentiate into osteoblasts and other cell types during development and regeneration. This is discussed in a larger context of regeneration in vertebrates as well as the clinical implication that this shift in understanding presents.
Collapse
Affiliation(s)
- R.S. Marcucio
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - T. Miclau
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - C.S. Bahney
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| |
Collapse
|
14
|
Novak S, Kalajzic I. AcanCreER lacks specificity to chondrocytes and targets periosteal progenitors in the fractured callus. Bone 2023; 166:116599. [PMID: 36309308 PMCID: PMC9832919 DOI: 10.1016/j.bone.2022.116599] [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: 09/26/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Aggrecan (Acan) is a large proteoglycan molecule constituting the extracellular matrix of cartilage, secreted by chondrocytes. To specifically target the chondrocyte lineage, researchers have widely used the AcanCreER mouse model. Evaluation of specificity and efficiency of recombination, requires Cre animals to be crossed with reporter mice. In order to accurately interpret data from Cre models, it is imperative to consider A) the amount of recombination occurring in cells/tissues that are not intended for targeting (i.e., non-specific expression), B) the efficiency of Cre recombination, which can depend on dose and duration of tamoxifen treatment, and C) the activation of CreER without tamoxifen induction, known as "Cre leakage." Using a highly sensitive reporter mouse (Ai9, tdTomato), we performed a comprehensive analysis of the AcanCreER system. Surprisingly, we observed expression in cells within the periosteum. These cells expand at a stage when chondrocytes are not yet present within the forming callus tissue (Acan/Ai9+ cells). In pulse-chase experiments, we confirmed that fibroblastic Acan/Ai9+ cells within the periosteum can directly give rise to osteoblasts. Our results show that Acan/Ai9+ is not specific for the chondrocyte lineage in the fracture callus or with the tibial holes. The expression of AcanCreER in periosteal progenitor cells complicates the interpretation of studies evaluating the transition of chondrocytes to osteoblasts (termed transdifferentiation). Awareness of these issues and the limitations of the system will lead to better data interpretation.
Collapse
Affiliation(s)
- Sanja Novak
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, USA.
| |
Collapse
|
15
|
Liu H, Tian F, Hu Y, Ping S, Zhang L. Liraglutide in Combination with Insulin Has a Superior Therapeutic Effect to Either Alone on Fracture Healing in Diabetic Rats. Diabetes Metab Syndr Obes 2023; 16:1235-1245. [PMID: 37151908 PMCID: PMC10155808 DOI: 10.2147/dmso.s404392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Purpose Fractures in patients with type 2 diabetes mellitus are at a high risk of delayed union or non-union. Previous studies have shown a protective effect of liraglutide on bone. In the present study, we aimed to investigate the effects of a combination of liraglutide and insulin on fracture healing in a rat model of diabetes and the mechanisms involved. Materials and Methods Closed femoral mid-shaft fractures were established in male Sprague-Dawley rats with or without diabetes mellitus, and the diabetic rats were administered insulin and/or liraglutide. Six weeks after femoral fracture, the femoral callus was evaluated by immunohistochemistry, histology, and micro-computed tomography. Additionally, the effects of liraglutide on high-glucose-stimulated MC3T3-E1 cells were analyzed by Western blotting. Results Micro-computed tomography and safranin O/fast green staining showed that fracture healing was delayed in the diabetic rats, and this was accompanied by much lower expression of osteogenic markers and greater osteoclast activity. However, treatment with insulin and/or liraglutide prevented these changes. Liraglutide in combination with insulin treatment resulted in lower blood glucose concentrations and significantly higher osteocalcin (OCN) and collagen I (Col I) expression six weeks following fracture. Western blot analysis showed that liraglutide prevented the low expression of the bone morphogenetic protein-2, osterix/SP7, OCN, Col I, and β-catenin in high-glucose-stimulated MC3T3-E1 cells. Conclusion These results demonstrate that insulin and/or liraglutide promotes bone fracture healing in the DF model. The combination was more effective than either single treatment, which may be because of the two drugs' additive effects on the osteogenic ability of osteoblast precursors.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
- Department of Orthopedic Syrgery, The Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei, People's Republic of China
| | - Faming Tian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, People’s Republic of China
| | - Yunpeng Hu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, People’s Republic of China
| | - Shaohua Ping
- Department of Orthopedic Syrgery, The Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei, People's Republic of China
| | - Liu Zhang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
- Department of Orthopedic Surgery, Emergency Management General Hospital, Beijing, People’s Republic of China
- Correspondence: Liu Zhang, Department of Orthopedic Surgery, Emergency Management General Hospital, Xibahenanli 29, Chaoyang dis, Beijing, 100028, People’s Republic of China, Tel +86-10-64662308, Email
| |
Collapse
|
16
|
The role of hypertrophic chondrocytes in regulation of the cartilage-to-bone transition in fracture healing. Bone Rep 2022; 17:101616. [PMID: 36105852 PMCID: PMC9465425 DOI: 10.1016/j.bonr.2022.101616] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/05/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Endochondral bone formation is an important pathway in fracture healing, involving the formation of a cartilaginous soft callus and the process of cartilage-to-bone transition. Failure or delay in the cartilage-to-bone transition causes an impaired bony union such as nonunion or delayed union. During the healing process, multiple types of cells including chondrocytes, osteoprogenitors, osteoblasts, and endothelial cells coexist in the callus, and inevitably crosstalk with each other. Hypertrophic chondrocytes located between soft cartilaginous callus and bony hard callus mediate the crosstalk regulating cell-matrix degradation, vascularization, osteoclast recruitment, and osteoblast differentiation in autocrine and paracrine manners. Furthermore, hypertrophic chondrocytes can become osteoprogenitors and osteoblasts, and directly contribute to woven bone formation. In this review, we focus on the roles of hypertrophic chondrocytes in fracture healing and dissect the intermingled crosstalk in fracture callus during the cartilage-to-bone transition.
Collapse
|
17
|
The Emerging Role of Cell Transdifferentiation in Skeletal Development and Diseases. Int J Mol Sci 2022; 23:ijms23115974. [PMID: 35682655 PMCID: PMC9180549 DOI: 10.3390/ijms23115974] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The vertebrate musculoskeletal system is known to be formed by mesenchymal stem cells condensing into tissue elements, which then differentiate into cartilage, bone, tendon/ligament, and muscle cells. These lineage-committed cells mature into end-stage differentiated cells, like hypertrophic chondrocytes and osteocytes, which are expected to expire and to be replaced by newly differentiated cells arising from the same lineage pathway. However, there is emerging evidence of the role of cell transdifferentiation in bone development and disease. Although the concept of cell transdifferentiation is not new, a breakthrough in cell lineage tracing allowed scientists to trace cell fates in vivo. Using this powerful tool, new theories have been established: (1) hypertrophic chondrocytes can transdifferentiate into bone cells during endochondral bone formation, fracture repair, and some bone diseases, and (2) tendon cells, beyond their conventional role in joint movement, directly participate in normal bone and cartilage formation, and ectopic ossification. The goal of this review is to obtain a better understanding of the key roles of cell transdifferentiation in skeletal development and diseases. We will first review the transdifferentiation of chondrocytes to bone cells during endochondral bone formation. Specifically, we will include the history of the debate on the fate of chondrocytes during bone formation, the key findings obtained in recent years on the critical factors and molecules that regulate this cell fate change, and the role of chondrocyte transdifferentiation in skeletal trauma and diseases. In addition, we will also summarize the latest discoveries on the novel roles of tendon cells and adipocytes on skeletal formation and diseases.
Collapse
|
18
|
Chen K, Henn D, Januszyk M, Barrera JA, Noishiki C, Bonham CA, Griffin M, Tevlin R, Carlomagno T, Shannon T, Fehlmann T, Trotsyuk AA, Padmanabhan J, Sivaraj D, Perrault DP, Zamaleeva AI, Mays CJ, Greco AH, Kwon SH, Leeolou MC, Huskins SL, Steele SR, Fischer KS, Kussie HC, Mittal S, Mermin-Bunnell AM, Diaz Deleon NM, Lavin C, Keller A, Longaker MT, Gurtner GC. Disrupting mechanotransduction decreases fibrosis and contracture in split-thickness skin grafting. Sci Transl Med 2022; 14:eabj9152. [PMID: 35584231 DOI: 10.1126/scitranslmed.abj9152] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Burns and other traumatic injuries represent a substantial biomedical burden. The current standard of care for deep injuries is autologous split-thickness skin grafting (STSG), which frequently results in contractures, abnormal pigmentation, and loss of biomechanical function. Currently, there are no effective therapies that can prevent fibrosis and contracture after STSG. Here, we have developed a clinically relevant porcine model of STSG and comprehensively characterized porcine cell populations involved in healing with single-cell resolution. We identified an up-regulation of proinflammatory and mechanotransduction signaling pathways in standard STSGs. Blocking mechanotransduction with a small-molecule focal adhesion kinase (FAK) inhibitor promoted healing, reduced contracture, mitigated scar formation, restored collagen architecture, and ultimately improved graft biomechanical properties. Acute mechanotransduction blockade up-regulated myeloid CXCL10-mediated anti-inflammation with decreased CXCL14-mediated myeloid and fibroblast recruitment. At later time points, mechanical signaling shifted fibroblasts toward profibrotic differentiation fates, and disruption of mechanotransduction modulated mesenchymal fibroblast differentiation states to block those responses, instead driving fibroblasts toward proregenerative, adipogenic states similar to unwounded skin. We then confirmed these two diverging fibroblast transcriptional trajectories in human skin, human scar, and a three-dimensional organotypic model of human skin. Together, pharmacological blockade of mechanotransduction markedly improved large animal healing after STSG by promoting both early, anti-inflammatory and late, regenerative transcriptional programs, resulting in healed tissue similar to unwounded skin. FAK inhibition could therefore supplement the current standard of care for traumatic and burn injuries.
Collapse
Affiliation(s)
- Kellen Chen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Surgery, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Dominic Henn
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Janos A Barrera
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chikage Noishiki
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Clark A Bonham
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ruth Tevlin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Theresa Carlomagno
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tara Shannon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Artem A Trotsyuk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jagannath Padmanabhan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dharshan Sivaraj
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David P Perrault
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alsu I Zamaleeva
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chyna J Mays
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Autumn H Greco
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sun Hyung Kwon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Melissa C Leeolou
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Savana L Huskins
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sydney R Steele
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katharina S Fischer
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hudson C Kussie
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Smiti Mittal
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alana M Mermin-Bunnell
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nestor M Diaz Deleon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christopher Lavin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany.,Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Geoffrey C Gurtner
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Surgery, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| |
Collapse
|
19
|
Kraus JM, Giovannone D, Rydzik R, Balsbaugh JL, Moss IL, Schwedler JL, Bertrand JY, Traver D, Hankenson KD, Crump JG, Youngstrom DW. Notch signaling enhances bone regeneration in the zebrafish mandible. Development 2022; 149:dev199995. [PMID: 35178545 PMCID: PMC8959151 DOI: 10.1242/dev.199995] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022]
Abstract
Loss or damage to the mandible caused by trauma, treatment of oral malignancies, and other diseases is treated using bone-grafting techniques that suffer from numerous shortcomings and contraindications. Zebrafish naturally heal large injuries to mandibular bone, offering an opportunity to understand how to boost intrinsic healing potential. Using a novel her6:mCherry Notch reporter, we show that canonical Notch signaling is induced during the initial stages of cartilage callus formation in both mesenchymal cells and chondrocytes following surgical mandibulectomy. We also show that modulation of Notch signaling during the initial post-operative period results in lasting changes to regenerate bone quantity one month later. Pharmacological inhibition of Notch signaling reduces the size of the cartilage callus and delays its conversion into bone, resulting in non-union. Conversely, conditional transgenic activation of Notch signaling accelerates conversion of the cartilage callus into bone, improving bone healing. Given the conserved functions of this pathway in bone repair across vertebrates, we propose that targeted activation of Notch signaling during the early phases of bone healing in mammals may both augment the size of the initial callus and boost its ossification into reparative bone.
Collapse
Affiliation(s)
- Jessica M. Kraus
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Dion Giovannone
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Renata Rydzik
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jeremy L. Balsbaugh
- Proteomics & Metabolomics Facility, Center for Open Research Resources & Equipment, University of Connecticut, Storrs, CT 06269, USA
| | - Isaac L. Moss
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jennifer L. Schwedler
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Julien Y. Bertrand
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - David Traver
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Daniel W. Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
20
|
Fernández-Iglesias Á, Fuente R, Gil-Peña H, Alonso-Durán L, Santos F, López JM. The Formation of the Epiphyseal Bone Plate Occurs via Combined Endochondral and Intramembranous-Like Ossification. Int J Mol Sci 2021; 22:ijms22020900. [PMID: 33477458 PMCID: PMC7830543 DOI: 10.3390/ijms22020900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 11/24/2022] Open
Abstract
The formation of the epiphyseal bone plate, the flat bony structure that provides strength and firmness to the growth plate cartilage, was studied in the present study by using light, confocal, and scanning electron microscopy. Results obtained evidenced that this bone tissue is generated by the replacement of the lower portion of the epiphyseal cartilage. However, this process differs considerably from the usual bone tissue formation through endochondral ossification. Osteoblasts deposit bone matrix on remnants of mineralized cartilage matrix that serve as a scaffold, but also on non-mineralized cartilage surfaces and as well as within the perivascular space. These processes occur simultaneously at sites located close to each other, so that, a core of the sheet of bone is established very quickly. Subsequently, thickening and reshaping occurs by appositional growth to generate a dense parallel-fibered bone structurally intermediate between woven and lamellar bone. All these processes occur in close relationship with a cartilage but most of the bone tissue is generated in a manner that may be considered as intramembranous-like. Overall, the findings here reported provide for the first time an accurate description of the tissues and events involved in the formation of the epiphyseal bone plate and gives insight into the complex cellular events underlying bone formation at different sites on the skeleton.
Collapse
Affiliation(s)
- Ángela Fernández-Iglesias
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Asturias, Spain; (Á.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (F.S.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Rocío Fuente
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Asturias, Spain; (Á.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (F.S.)
| | - Helena Gil-Peña
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Asturias, Spain; (Á.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (F.S.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
- Department of Pediatrics, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Asturias, Spain
| | - Laura Alonso-Durán
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Asturias, Spain; (Á.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (F.S.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Fernando Santos
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Asturias, Spain; (Á.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (F.S.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
- Department of Pediatrics, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Asturias, Spain
| | - José Manuel López
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Asturias, Spain; (Á.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (F.S.)
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Asturias, Spain
- Correspondence:
| |
Collapse
|