1
|
Nafisi N, Razavi AH, Shariyate MJ, Velasquez MV, Khak M, Manoukian D, Klujian A, Mirzamohammadi H, Cummiskey T, Rostami MR, Mirzamohammadi F, Nazarian A. Decoding fracture healing: A scoping review of mechanistic pathways derived from transcriptional analysis in murine studies. Bone 2025; 194:117444. [PMID: 40032014 DOI: 10.1016/j.bone.2025.117444] [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: 01/24/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Fracture healing is a complex biological process involving orchestrated interactions among cells, growth factors, and transcriptional pathways. Despite significant advancements in understanding bone repair, non-union and delayed healing remain prevalent, especially in patients with comorbidities such as aging, diabetes, or substance use. Murine models serve as a cornerstone in fracture healing research, offering genetic manipulability, cost-effectiveness, and biological relevance to humans. This scoping review consolidates findings from studies conducted between 2010 and 2024, focusing on mechanistic pathways derived from transcriptional analysis in secondary bone healing as identified through bulk RNA sequencing of murine models. Key mechanistic pathways were categorized and analyzed across the distinct phases of fracture healing-reactive, reparative, and remodeling-highlighting their unique roles in inflammation, ECM remodeling, cell proliferation, and tissue mineralization. The most recurrent mechanistic pathways included ECM-receptor interaction, focal adhesion, and signaling mechanisms such as MAPK and TGF-beta. Variability in methodologies and limited overlap among studies underscore the need for standardized protocols in RNA sequencing analysis. Additionally, comparisons across long bone fractures, hole defects, and craniofacial bone healing revealed shared molecular mechanisms alongside unique challenges, particularly in craniofacial models. This scoping review underscores the promise of integrating systems biology approaches with transcriptomic data to elucidate the intricate regulatory networks governing fracture repair. Addressing the identified gaps in early-phase healing and harmonizing research methodologies will advance therapeutic strategies to reduce non-union rates and improve clinical outcomes.
Collapse
Affiliation(s)
- Nazanin Nafisi
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Ahmad Hedayatzadeh Razavi
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Mohammad Javad Shariyate
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maria V Velasquez
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mohammad Khak
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Manoukian
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Arthur Klujian
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hamid Mirzamohammadi
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medica Sciences, Tehran, Iran
| | - Tom Cummiskey
- Knowledge Services, Beth Israel Lahey Health, Cambridge, MA, USA
| | | | | | - Ara Nazarian
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Mechanical Engineering, Boston University, Boston, MA, USA; Department of Orthopaedic Surgery, Yerevan State Medical University, Yerevan, Armenia.
| |
Collapse
|
2
|
Liu F, Hu Y, Zhang Y, Ren C, Qiao F, Yang H, Xu H, Yang P. Aging-Induced Discrepant Response of Fracture Healing is Initiated from the Organization and Mineralization of Collagen Fibrils in Callus. ACS Biomater Sci Eng 2025; 11:1038-1050. [PMID: 39831893 DOI: 10.1021/acsbiomaterials.4c01490] [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: 01/22/2025]
Abstract
Fracture healing is a complex process during which the bone restores its structural and mechanical integrity. Collagen networks and minerals are the fundamental components to rebuild the bone matrix in callus. It has been recognized that bone quality could be impaired during aging. However, how the structural and mechanical recovery of fracture healing is influenced by aging, particularly from the perspective of organization and mineralization of the collagen network in callus, remains unclear. A tibial fracture model was established for both the young (5 weeks) and aged mice (68 weeks). On the 21st day postfracture, the characteristics of the collagen network, mineralization, and the nanoscale mechanical properties of the callus were assessed. The results indicated that aging postpones the fracture healing process, leading to incomplete microstructure, less mineral content and mineralization, and weaker mechanical properties of callus. In the aged mice, the internal fixation and mechanical immobilization promoted the mineralization of callus by increasing mineral crystal length and mineral-to-matrix ratio by 48 and 42% compared to the internal fixation and free movement control group, respectively. By contrast, in the young mice, the internal fixation and mechanical immobilization induced disordered collagen fibrils and decreased the crystal length and mineral-to-matrix ratio by 32 and 36%, compared to the internal fixation and free movement control group, respectively. The present findings suggested that the aging-induced structure and mechanical differences of callus during fracture healing initiate from the organization and mineralization of collagen fibrils. Multiscale structural and mechanical analysis suggested mechanical immobilization is beneficial to the structure, composition, and mechanics of callus in the aged mice while impairing the organization and mineralization of collagen fibril in the callus of the young mice. These findings suggested that different mechanical intervention strategies should be adopted for fracture healing at different ages, which provides valuable insights for the clinical treatment of bone fracture.
Collapse
Affiliation(s)
- Fa Liu
- Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Ningbo 315103, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yiwei Hu
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yuzhi Zhang
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Chenxi Ren
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Feng Qiao
- Department of Orthopedics Combined TCM with Western Medicine, Honghui Hospital, Xi'an Jiaotong University, No. 555 Youyi East Road, Xi'an, Shaanxi 710054, China
| | - Hui Yang
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Huiyun Xu
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Pengfei Yang
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
3
|
Jörimann T, Füllemann P, Jose A, Matthys R, Wehrle E, Stoddart MJ, Verrier S. In Vitro Induction of Hypertrophic Chondrocyte Differentiation of Naïve MSCs by Strain. Cells 2024; 14:25. [PMID: 39791725 PMCID: PMC11720650 DOI: 10.3390/cells14010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
In the context of bone fractures, the influence of the mechanical environment on the healing outcome is widely accepted, while its influence at the cellular level is still poorly understood. This study explores the influence of mechanical load on naïve mesenchymal stem cell (MSC) differentiation, focusing on hypertrophic chondrocyte differentiation. Unlike primary bone healing, which involves the direct differentiation of MSCs into bone-forming cells, endochondral ossification uses an intermediate cartilage template that remodels into bone. A high-throughput uniaxial bioreactor system (StrainBot) was used to apply varying percentages of strain on naïve MSCs encapsulated in GelMa hydrogels. This research shows that cyclic uniaxial compression alone directs naïve MSCs towards a hypertrophic chondrocyte phenotype. This was demonstrated by increased cell volumes and reduced glycosaminoglycan (GAG) production, along with an elevated expression of hypertrophic markers such as MMP13 and Type X collagen. In contrast, Type II collagen, typically associated with resting chondrocytes, was poorly detected under mechanical loading alone conditions. The addition of chondrogenic factor TGFβ1 in the culture medium altered these outcomes. TGFβ1 induced chondrogenic differentiation, as indicated by higher GAG/DNA production and Type II collagen expression, overshadowing the effect of mechanical loading. This suggests that, under mechanical strain, hypertrophic differentiation is hindered by TGFβ1, while chondrogenesis is promoted. Biochemical analyses further confirmed these findings. Mechanical deformation alone led to a larger cell size and a more rounded cell morphology characteristic of hypertrophic chondrocytes, while lower GAG and proteoglycan production was observed. Immunohistology staining corroborated the gene expression data, showing increased Type X collagen with mechanical strain. Overall, this study indicates that mechanical loading alone drives naïve MSCs towards a hypertrophic chondrocyte differentiation path. These insights underscore the critical role of mechanical forces in MSC differentiation and have significant implications for bone healing, regenerative medicine strategies and rehabilitation protocols.
Collapse
Affiliation(s)
- Thomas Jörimann
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland (E.W.); (M.J.S.)
| | - Priscilla Füllemann
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland (E.W.); (M.J.S.)
| | - Anita Jose
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland (E.W.); (M.J.S.)
| | | | - Esther Wehrle
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland (E.W.); (M.J.S.)
| | - Martin J. Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland (E.W.); (M.J.S.)
| | - Sophie Verrier
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland (E.W.); (M.J.S.)
| |
Collapse
|
4
|
Wen J, Zeng Y, Su S, Song M, Wang Z, Chen X, Dong X. Magnesium degradation-induced variable fixation plates promote bone healing in rabbits. J Orthop Traumatol 2024; 25:56. [PMID: 39572420 PMCID: PMC11582307 DOI: 10.1186/s10195-024-00803-0] [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: 08/01/2024] [Accepted: 11/03/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Both initial mechanical stability and subsequent axial interfragmentary micromotion at fracture ends play crucial roles in fracture healing. However, the conversion timing of variable fixation and its effect on and mechanism of fracture healing remain inadequately explored. METHODS A magnesium degradation-induced variable fixation plate (MVFP) for femurs was designed, and its conversion timing was investigated both in vitro and in vivo. Then, locking plates and MVFPs with and without a magnesium shim were implanted in rabbit femur fracture models. X-ray photography and micro computed tomography (micro-CT) were performed to observe the healing of the fracture. Toluidine blue and Masson's trichrome staining were performed to observe new bone formation. The torsion test was used to determine the strength of the bone after healing. Finally, reverse transcription-polymerase chain reaction (RT-PCR) and western blotting were used to detect the expression of osteogenesis-related genes in the three groups. RESULTS The MVFP with sample 3 magnesium shim showed greater axial displacement within 15 days in vitro, and its variable capability was likewise confirmed in vivo. X-ray photography and micro-CT indicated increased callus formation in the variable fixation group. Toluidine blue and Masson's trichrome staining revealed less callus formation on the rigid fixation side of the locking plate, whereas the variable fixation group presented more callus formation, more symmetrical intraosseous calli, and greater maturity. The torsion test indicated greater torsional resistance of the healed bone in the variable fixation group. RT-PCR and western blotting revealed that the expression levels of BMP2 and OPG increased during early fracture stages but decreased in late fracture stages, whereas RANKL expression showed the opposite trend in the variable fixation group. CONCLUSIONS MVFP promoted faster and stronger bone healing in rabbits, potentially by accelerating the expression of BMP2 and modulating the OPG/RANKL/RANK signaling axis. This study offers valuable insights for the clinical application of variable fixation technology in bone plates and contributes to the advancement of both internal fixation technology and theory. LEVEL OF EVIDENCE level V.
Collapse
Affiliation(s)
- Jian Wen
- JXHC Key Laboratory of Digital Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
- Department of Pain Management, The 2Nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Yu Zeng
- JXHC Key Laboratory of Digital Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
- Jiangxi University of Chinese Medicine, No. 1688, Meiling Avenue, Nanchang, 330004, Jiangxi, China
| | - Shenghui Su
- JXHC Key Laboratory of Digital Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Meiling Song
- JXHC Key Laboratory of Digital Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
- Ruijin Traditional Chinese Medicine Hospital, Ruijin, 342500, Jiangxi, China
| | - Zhe Wang
- JXHC Key Laboratory of Digital Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Xiaofan Chen
- JXHC Key Laboratory of Digital Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Xieping Dong
- JXHC Key Laboratory of Digital Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
5
|
Shi V, Morgan EF. Estrogen and estrogen receptors mediate the mechanobiology of bone disease and repair. Bone 2024; 188:117220. [PMID: 39106937 PMCID: PMC11392539 DOI: 10.1016/j.bone.2024.117220] [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: 06/08/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
It is well understood that the balance of bone formation and resorption is dependent on both mechanical and biochemical factors. In addition to cell-secreted cytokines and growth factors, sex hormones like estrogen are critical to maintaining bone health. Although the direct osteoprotective function of estrogen and estrogen receptors (ERs) has been reported extensively, evidence that estrogen signaling also has a role in mediating the effects of mechanical loading on maintenance of bone mass and healing of bone injuries has more recently emerged. Recent studies have underscored the role of estrogen and ERs in many pathways of bone mechanosensation and mechanotransduction. Estrogen and ERs have been shown to augment integrin-based mechanotransduction as well as canonical Wnt/b-catenin, RhoA/ROCK, and YAP/TAZ pathways. Estrogen and ERs also influence the mechanosensitivity of not only osteocytes but also osteoblasts, osteoclasts, and marrow stromal cells. The current review will highlight these roles of estrogen and ERs in cellular mechanisms underlying bone mechanobiology and discuss their implications for management of osteoporosis and bone fractures. A greater understanding of the mechanisms behind interactions between estrogen and mechanical loading may be crucial to addressing the shortcomings of current hormonal and pharmaceutical therapies. A combined therapy approach including high-impact exercise therapy may mitigate adverse side effects and allow an effective long-term solution for the prevention, treatment, and management of bone fragility in at-risk populations. Furthermore, future implications to novel local delivery mechanisms of hormonal therapy for osteoporosis treatment, as well as the effects on bone health of applications of sex hormone therapy outside of bone disease, will be discussed.
Collapse
Affiliation(s)
- Vivian Shi
- Boston University, Department of Biomedical Engineering, 44 Cummington St, Boston 02215, MA, USA; Center for Multiscale and Translational Mechanobiology, Boston University, 44 Cummington St, Boston 02215, MA, USA
| | - Elise F Morgan
- Boston University, Department of Biomedical Engineering, 44 Cummington St, Boston 02215, MA, USA; Center for Multiscale and Translational Mechanobiology, Boston University, 44 Cummington St, Boston 02215, MA, USA.
| |
Collapse
|
6
|
Wang Z, Lin M, Pan Y, Liu Y, Yang C, Wu J, Wang Y, Yan B, Zhou J, Chen R, Liu C. Periostin + myeloid cells improved long bone regeneration in a mechanosensitive manner. Bone Res 2024; 12:59. [PMID: 39406726 PMCID: PMC11480347 DOI: 10.1038/s41413-024-00361-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 07/06/2024] [Accepted: 08/01/2024] [Indexed: 10/19/2024] Open
Abstract
Myeloid cells are pivotal in the inflammatory and remodeling phases of fracture repair. Here, we investigate the effect of periostin expressed by myeloid cells on bone regeneration in a monocortical tibial defect (MTD) model. In this study, we show that periostin is expressed by periosteal myeloid cells, primarily the M2 macrophages during bone regeneration. Knockout of periostin in myeloid cells reduces cortical bone thickness, disrupts trabecular bone connectivity, impairs repair impairment, and hinders M2 macrophage polarization. Mechanical stimulation is a regulator of periostin in macrophages. By activating transforming growth factor-β (TGF-β), it increases periostin expression in macrophages and induces M2 polarization. This mechanosensitive effect also reverses the delayed bone repair induced by periostin deficiency in myeloid cells by strengthening the angiogenesis-osteogenesis coupling. In addition, transplantation of mechanically conditioned macrophages into the periosteum over a bone defect results in substantially enhanced repair, confirming the critical role of macrophage-secreted periostin in bone repair. In summary, our findings suggest that mechanical stimulation regulates periostin expression and promotes M2 macrophage polarization, highlighting the potential of mechanically conditioned macrophages as a therapeutic strategy for enhancing bone repair.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Minmin Lin
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yonghao Pan
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yang Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Chengyu Yang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Jianqun Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Bingtong Yan
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Jingjing Zhou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Rouxi Chen
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
7
|
VAN Hooren B, VAN Rengs L, Meijer K. Predicting Musculoskeletal Loading at Common Running Injury Locations Using Machine Learning and Instrumented Insoles. Med Sci Sports Exerc 2024; 56:2059-2075. [PMID: 38857523 DOI: 10.1249/mss.0000000000003493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
INTRODUCTION Wearables have the potential to provide accurate estimates of tissue loads at common running injury locations. Here we investigate the accuracy by which commercially available instrumented insoles (ARION; ATO-GEAR, Eindhoven, The Netherlands) can predict musculoskeletal loading at common running injury locations. METHODS Nineteen runners (10 males) ran at five different speeds, four slopes, with different step frequencies, and forward trunk lean on an instrumented treadmill while wearing instrumented insoles. The insole data were used as input to an artificial neural network that was trained to predict the Achilles tendon strain, and tibia and patellofemoral stress impulses and weighted impulses (damage proxy) as determined with musculoskeletal modeling. Accuracy was investigated using leave-one-out cross-validation and correlations. The effect of different input metrics was also assessed. RESULTS The neural network predicted tissue loading with overall relative percentage errors of 1.95 ± 8.40%, -7.37 ± 6.41%, and -12.8 ± 9.44% for the patellofemoral joint, tibia, and Achilles tendon impulse, respectively. The accuracy significantly changed with altered running speed, slope, or step frequency. Mean (95% confidence interval) within-individual correlations between modeled and predicted impulses across conditions were generally nearly perfect, being 0.92 (0.89 to 0.94), 0.95 (0.93 to 0.96), and 0.95 (0.94 to 0.96) for the patellofemoral, tibial, and Achilles tendon stress/strain impulses, respectively. CONCLUSIONS This study shows that commercially available instrumented insoles can predict loading at common running injury locations with variable absolute but (very) high relative accuracy. The absolute error was lower than the methods that measure only the step count or assume a constant load per speed or slope. This developed model may allow for quantification of in-field tissue loading and real-time tissue loading-based feedback to reduce injury risk.
Collapse
Affiliation(s)
- Bas VAN Hooren
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Department of Nutrition and Movement Sciences, Maastricht, THE NETHERLANDS
| | | | | |
Collapse
|
8
|
Bowers KM, Anderson DE. Delayed Union and Nonunion: Current Concepts, Prevention, and Correction: A Review. Bioengineering (Basel) 2024; 11:525. [PMID: 38927761 PMCID: PMC11201148 DOI: 10.3390/bioengineering11060525] [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: 03/29/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Surgical management of fractures has advanced with the incorporation of advanced technology, surgical techniques, and regenerative therapies, but delayed bone healing remains a clinical challenge and the prevalence of long bone nonunion ranges from 10 to 15% of surgically managed fractures. Delayed bone healing arises from a combination of mechanical, biological, and systemic factors acting on the site of tissue remodeling, and careful consideration of each case's injury-related, patient-dependent, surgical, and mechanical risk factors is key to successful bone union. In this review, we describe the biology and biomechanics of delayed bone healing, outline the known risk factors for nonunion development, and introduce modern preventative and corrective therapies targeting fracture nonunion.
Collapse
Affiliation(s)
| | - David E. Anderson
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, 2407 River Dr., Knoxville, TN 37996-4550, USA;
| |
Collapse
|
9
|
Wähnert D, Miersbach M, Colcuc C, Brianza S, Vordemvenne T, Plecko M, Schwarz A. Promoting bone callus formation by taking advantage of the time-dependent fracture gap strain modulation. Front Surg 2024; 11:1376441. [PMID: 38756355 PMCID: PMC11096559 DOI: 10.3389/fsurg.2024.1376441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
Delayed union and non-union of fractures continue to be a major problem in trauma and orthopedic surgery. These cases are challenging for the surgeon. In addition, these patients suffer from multiple surgeries, pain and disability. Furthermore, these cases are a major burden on healthcare systems. The scientific community widely agrees that the stability of fixation plays a crucial role in determining the outcome of osteosynthesis. The extent of stabilization affects factors like fracture gap strain and fluid flow, which, in turn, influence the regenerative processes positively or negatively. Nonetheless, a growing body of literature suggests that during the fracture healing process, there exists a critical time frame where intervention can stimulate the bone's return to its original form and function. This article provides a summary of existing evidence in the literature regarding the impact of different levels of fixation stability on the strain experienced by newly forming tissues. We will also discuss the timing and nature of this "window of opportunity" and explore how current knowledge is driving the development of new technologies with design enhancements rooted in mechanobiological principles.
Collapse
Affiliation(s)
- Dirk Wähnert
- Department of Trauma and Orthopedic Surgery, Protestant Hospital of Bethel Foundation, University Hospital OWL of Bielefeld University, Bielefeld, Germany
| | - Marco Miersbach
- Department of Trauma and Orthopedic Surgery, Protestant Hospital of Bethel Foundation, University Hospital OWL of Bielefeld University, Bielefeld, Germany
| | - Christian Colcuc
- Department of Trauma and Orthopedic Surgery, Protestant Hospital of Bethel Foundation, University Hospital OWL of Bielefeld University, Bielefeld, Germany
| | | | - Thomas Vordemvenne
- Department of Trauma and Orthopedic Surgery, Protestant Hospital of Bethel Foundation, University Hospital OWL of Bielefeld University, Bielefeld, Germany
| | - Michael Plecko
- Department of Orthopaedics and Traumatology, Trauma Hospital Graz (UKH), Graz, Austria
| | - Angelika Schwarz
- Department of Orthopaedics and Traumatology, Trauma Hospital Graz (UKH), Graz, Austria
| |
Collapse
|
10
|
Takaoka Y, Fujibayashi S, Onoe H, Goto K, Otsuki B, Kawai T, Okuzu Y, Shimizu T, Ikeda N, Orita K, Honda S, Ikezaki T, Yabutsuka T, Matsuda S. Bone ingrowth into a porous structure is achieved by preceding fibrogenesis and vascularization. Acta Biomater 2024; 177:243-252. [PMID: 38367656 DOI: 10.1016/j.actbio.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/13/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Porous structures are frequently used in surgical implants to strengthen the interlocking power produced by bone ingrowth. Therefore, we aimed to elucidate the mechanism underlying bone ingrowth into a porous structure accompanied by vascularization. A nonbioactive polyetheretherketone implant with a 3D-printed porous structure was prepared and implanted in a bone hole created in the tibias of rabbits. We observed bone ingrowth in the same individual specimens immediately and at 2, 4, 8, and 12 weeks post-implantation using in-vivo computed tomography (CT). Furthermore, a detailed evaluation with blood vessels of each specimen at 2, 4, and 12 weeks was performed with ex-vivo CT and histological specimen. Additional histological evaluation was performed using thin sections of an implant made with thermoplastic polyurethane having the same structure. As a result, the bone invasion began after four weeks, when the construction of fibrous tissue and the spread of new blood vessels within the voids matured. As the bone matured in the load-bearing area, new blood vessels outside the bone matrix regressed. This longitudinal evaluation study suggests that preceding fibrogenesis and vascularization may be key in developing bone ingrowth. STATEMENT OF SIGNIFICANCE: A porous structure is an essential structure for dental and orthopedic implants because it provides strong fixation through bone invasion. Although it was known that vascularization was involved in this, the details were not known. This in vivo study revealed that in order for bone ingrowth to begin, a preparatory period of approximately 4 weeks was required to establish blood flow inside and outside the implant. Furthermore, it was confirmed that by spreading the fibrous structure in advance, it has an advantageous effect on the migration of cells involved in the formation of bones and blood vessels. We pointed out that it is necessary to consider fibrogenesis and vascularization when creating future implants.
Collapse
Affiliation(s)
- Yusuke Takaoka
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan; Tenri Hospital, Nara, Japan.
| | - Shunsuke Fujibayashi
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Hirotaka Onoe
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Goto
- Department of Orthopaedic Surgery, Kindai University, Osaka, Japan
| | - Bungo Otsuki
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Toshiyuki Kawai
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Yaichiro Okuzu
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Takayoshi Shimizu
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Norimasa Ikeda
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Kazuki Orita
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Shintaro Honda
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Tatsuhito Ikezaki
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| | - Takeshi Yabutsuka
- Department of Fundamental Energy Science, Graduate School of Energy Science, Kyoto University, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto 606-8507, Japan
| |
Collapse
|
11
|
Li Z, Pollard S, Smith G, Deshmukh S, Ding Z. Biomechanical analysis of combi-hole locking compression plate during fracture healing: A numerical study of screw configuration. Proc Inst Mech Eng H 2024; 238:313-323. [PMID: 38372206 PMCID: PMC10941711 DOI: 10.1177/09544119241229157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/12/2024] [Indexed: 02/20/2024]
Abstract
Locking compression plates (LCPs) have become a widely used option for treating femur bone fractures. However, the optimal screw configuration with combi-holes remains a subject of debate. The study aims to create a time-dependent finite element (FE) model to assess the impacts of different screw configurations on LCP fixation stiffness and healing efficiency across four healing stages during a complete fracture healing process. To simulate the healing process, we integrated a time-dependent callus formation mechanism into a FE model of the LCP with combi-holes. Three screw configuration parameters, namely working length, screw number, and screw position, were investigated. Increasing the working length negatively affected axial stiffness and healing efficiency (p < 0.001), while screw number or position had no significant impact (p > 0.01). The time-dependent model displayed a moderate correlation with the conventional time-independent model for axial stiffness and healing efficiency (ρ ≥ 0.733, p ≤ 0.025). The highest healing efficiency (95.2%) was observed in screw configuration C125 during the 4-8-week period. The results provide insights into managing fractures using LCPs with combi-holes over an extended duration. Under axial compressive loading conditions, the use of the C125 screw configuration can enhance callus formation during the 4-12-week period for transverse fractures. When employing the C12345 configuration, it becomes crucial to avoid overconstraint during the 4-8-week period.
Collapse
Affiliation(s)
- Zeyang Li
- School of Engineering, Cardiff University, Cardiff, UK
| | - Stuart Pollard
- School of Engineering, University of Birmingham, Birmingham, UK
| | | | | | - Ziyun Ding
- School of Engineering, University of Birmingham, Birmingham, UK
| |
Collapse
|
12
|
Hu Y, Lian Q, Cao F, Hou X, Li H, Xing L, Wang M, Tian F, Zhang L. Estrogen deficiency impedes fracture healing despite eliminating the excessive absorption of the posterior callus in a semi-fixed distal tibial fracture mouse model. BMC Musculoskelet Disord 2023; 24:803. [PMID: 37817119 PMCID: PMC10563296 DOI: 10.1186/s12891-023-06929-2] [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: 04/06/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Treatment of distal tibial fractures is a challenge due to their specific anatomical location. However, there is no appropriate mouse model to simulate a clinical distal tibial fracture for basic research. The aim of this investigation was to evaluate the feasibility of simulating a clinical fracture of the distal tibia of mice and to investigate the effect of ovariectomy (OVX)-induced osteoporosis on fracture healing in this model. METHODS Sixty female 8-week-old C57BL/6 mice were randomly divided into two groups, either sham or OVX. A semi-fixation distal tibia fracture was established in the right tibia after 8 weeks of OVX. The right tibias were collected at 7, 14, 21, and 28 days post fracture. RESULTS In the semi-fixation distal tibia fracture model, the posterior callus in the sham group showed excessive bone resorption and lower bone mass phenotype compared with the anterior site; a similar trend was not found in the OVX group. At 28 days post fracture, the posterior callus was more mineralized than the anterior callus in the OVX group. Although the fracture healing of the sham group showed a special phenotype in this mode, the progress and quality of fracture healing were still better than those of the OVX group. CONCLUSION A semi-fixed distal tibial closed fracture mouse model was successfully established. In this model, excess bone resorption of the posterior callus impaired normal fracture healing, but not in OVX-induced osteoporotic bone. Although the stress shielding effect was not observed in the OVX group, impaired bone healing caused by OVX was still present. Our results suggest that this fracture model may have potential for studies on distal tibial fractures and stress shielding.
Collapse
Affiliation(s)
- Yunpeng Hu
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Qiangqiang Lian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, P. R. China
| | - Fuyuan Cao
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, P. R. China
| | - Xiaoli Hou
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, P. R. China
| | - Hetong Li
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Lei Xing
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, P. R. China
| | - Mengqin Wang
- Emergency Department, JST, The Fourth Clinical Hospital of Peking University, Beijing, P. R. China
| | - Faming Tian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, P. R. China.
| | - Liu Zhang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, Hebei, P. R. China.
- Department of Orthopedic Surgery, Emergency General Hospital, Xibahenanli29, Chaoyang Dis, Beijing, 100028, P. R. China.
| |
Collapse
|
13
|
Mi C, Zhang X, Yang C, Wu J, Chen X, Ma C, Wu S, Yang Z, Qiao P, Liu Y, Wu W, Guo Z, Liao J, Zhou J, Guan M, Liang C, Liu C, Jin D. Bone disease imaging through the near-infrared-II window. Nat Commun 2023; 14:6287. [PMID: 37813832 PMCID: PMC10562434 DOI: 10.1038/s41467-023-42001-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
Skeletal disorders are commonly diagnosed by X-ray imaging, but the radiation limits its use. Optical imaging through the near-infrared-II window (NIR-II, 1000-1700 nm) can penetrate deep tissues without radiation risk, but the targeting of contrast agent is non-specific. Here, we report that lanthanide-doped nanocrystals can passively target the bone marrow, which can be effective for over two months. We therefore develop the high-resolution NIR-II imaging method for bone disease diagnosis, including the 3D bone imaging instrumentation to show the intravital bone morphology. We demonstrate the monitoring of 1 mm bone defects with spatial resolution comparable to the X-ray imaging result. Moreover, NIR-II imaging can reveal the early onset inflammation as the synovitis in the early stage of rheumatoid arthritis, comparable to micro computed tomography (μCT) in diagnosis of osteoarthritis, including the symptoms of osteophyte and hyperostosis in the knee joint.
Collapse
Affiliation(s)
- Chao Mi
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
- National Institute of Extremely-Weak Magnetic Field Infrastructure, Hangzhou, China.
- Shenzhen Light Life Technology Co., Ltd., Shenzhen, China.
| | - Xun Zhang
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chengyu Yang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jianqun Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Chenguang Ma
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Sitong Wu
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Zhichao Yang
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Pengzhen Qiao
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Weijie Wu
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Zhiyong Guo
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- National Institute of Extremely-Weak Magnetic Field Infrastructure, Hangzhou, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Jiayan Liao
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Jiajia Zhou
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Ming Guan
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Light Life Technology Co., Ltd., Shenzhen, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China.
| | - Dayong Jin
- UTS-SUSTech Joint Research Centre for Biomedical Materials and Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
14
|
Synergistic effect of sulfonation followed by precipitation of amorphous calcium phosphate on the bone-bonding strength of carbon fiber reinforced polyetheretherketone. Sci Rep 2023; 13:1443. [PMID: 36697480 PMCID: PMC9876887 DOI: 10.1038/s41598-023-28701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/23/2023] [Indexed: 01/26/2023] Open
Abstract
Sulfonation and applications of amorphous calcium phosphate are known to make polyetheretherketone (PEEK) bioactive. Sulfonation followed by precipitation of amorphous calcium phosphate (AN-treatment) may provide PEEK with further bone-bonding strength. Herein, we prepared a carbon-fiber-reinforced PEEK (CPEEK) with similar tensile strength to cortical bone and a CPEEK subjected to AN-treatment (CPEEK-AN). The effect of AN-treatment on the bone-bonding strength generated at the interface between the rabbit's tibia and a base material was investigated using a detaching test at two time-points (4 and 8 weeks). At 4 weeks, the strength of CPEEK-AN was significantly higher than that of CPEEK due to the direct bonding between the interfaces. Between 4 and 8 weeks, the different bone forming processes showed that, with CPEEK-AN, bone consolidation was achieved, thus improving bone-bonding strength. In contrast, with CPEEK, a new bone was absorbed mainly on the interface, leading to poor strength. These observations were supported by an in vitro study, which showed that pre-osteoblast on CPEEK-AN caused earlier maturation and mineralization of the extracellular matrix than on CPEEK. Consequently, AN-treatment, comprising a combination of two efficient treatments, generated a synergetic effect on the bonding strength of CPEEK.
Collapse
|
15
|
Ma Q, Miri Z, Haugen HJ, Moghanian A, Loca D. Significance of mechanical loading in bone fracture healing, bone regeneration, and vascularization. J Tissue Eng 2023; 14:20417314231172573. [PMID: 37251734 PMCID: PMC10214107 DOI: 10.1177/20417314231172573] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
In 1892, J.L. Wolff proposed that bone could respond to mechanical and biophysical stimuli as a dynamic organ. This theory presents a unique opportunity for investigations on bone and its potential to aid in tissue repair. Routine activities such as exercise or machinery application can exert mechanical loads on bone. Previous research has demonstrated that mechanical loading can affect the differentiation and development of mesenchymal tissue. However, the extent to which mechanical stimulation can help repair or generate bone tissue and the related mechanisms remain unclear. Four key cell types in bone tissue, including osteoblasts, osteoclasts, bone lining cells, and osteocytes, play critical roles in responding to mechanical stimuli, while other cell lineages such as myocytes, platelets, fibroblasts, endothelial cells, and chondrocytes also exhibit mechanosensitivity. Mechanical loading can regulate the biological functions of bone tissue through the mechanosensor of bone cells intraosseously, making it a potential target for fracture healing and bone regeneration. This review aims to clarify these issues and explain bone remodeling, structure dynamics, and mechano-transduction processes in response to mechanical loading. Loading of different magnitudes, frequencies, and types, such as dynamic versus static loads, are analyzed to determine the effects of mechanical stimulation on bone tissue structure and cellular function. Finally, the importance of vascularization in nutrient supply for bone healing and regeneration was further discussed.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
- Department of Immunology, School of
Basic Medicine, Fourth Military Medical University, Xi’an, PR China
| | - Zahra Miri
- Department of Materials Engineering,
Isfahan University of Technology, Isfahan, Iran
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
| | - Amirhossein Moghanian
- Department of Materials Engineering,
Imam Khomeini International University, Qazvin, Iran
| | - Dagnjia Loca
- Rudolfs Cimdins Riga Biomaterials
Innovations and Development Centre, Institute of General Chemical Engineering,
Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga,
Latvia
- Baltic Biomaterials Centre of
Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
16
|
Ismiarto YD, Prasetiyo GT, Putra EP, Prasetya WP. Aseptic non-union fracture of ulna but union fracture of radius following internal fixation procedure: A case report. INTERNATIONAL JOURNAL OF SURGERY OPEN 2022. [DOI: 10.1016/j.ijso.2022.100540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Yang C, Liu Y, Wang Z, Lin M, Liu C. Controlled mechanical loading improves bone regeneration by regulating type H vessels in a S1Pr1-dependent manner. FASEB J 2022; 36:e22530. [PMID: 36063128 DOI: 10.1096/fj.202200339rrr] [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: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair and result in delayed unions or non-unions. Dynamic mechanical stimulation promotes bone formation, when applied at the correct time frame, with optimal loading magnitude, frequency, and repetition. Controlled mechanical loading significantly increases osteogenic cells during the matrix deposition phase of bone repair. In the bone defect, the blood vessel network guides the initial bone formation activities. A unique blood vessel subtype (Type H) exists in bone, which expresses high levels of CD31 and endomucin, and functions to couple angiogenesis and osteogenesis. However, how this form of controlled mechanical loading regulates the Type H vessels and promotes bone formation is still not clear. Sphingosine 1-phosphate (S1P) participates in the bone anabolic process and is a key regulator of the blood vessel. Its receptor, sphingosine 1-phosphate receptor 1 (S1Pr1), is a mechanosensitive protein that regulates vascular integrity. Therefore, we hypothesis that controlled anabolic mechanical loading promotes bone repair by acting on Type H vessels. To study the effect of S1Pr1 on loading induced-bone repair, we utilized a stabilized tibial defect model, which allows for the application of anabolic mechanical loading. Mechanical loading upregulated S1Pr1 within the entire defect, with up to 80% expressed in blood vessels, as observed by deep tissue imaging. Additionally, S1Pr1 antagonism by W146 inhibited the anabolic effects of mechanical loading. We showed that mechanical loading or activating S1Pr1 could induce YAP nuclear translocation, a key regulator in the cell's mechanical response, in endothelial cells (ECs) in vitro. Inhibition of S1Pr1 in endothelial cells by siRNA reduced loading-induced YAP nuclear translocation and expressions of angiogenic genes. In vivo, YAP nuclear translocation in Type H vessels was up-regulated after mechanical loading but was inhibited by antagonizing S1Pr1. S1Pr1 agonist, FTY720, increased bone volume and Type H vessel volume, similar to that of mechanical stimulation. In conclusion, controlled anabolic mechanical loading enhanced bone formation mainly through Type H vessels in a S1Pr1-dependent manner.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
18
|
Yang C, Li Z, Liu Y, Hou R, Lin M, Fu L, Wu D, Liu Q, Li K, Liu C. Single-cell spatiotemporal analysis reveals cell fates and functions of transplanted mesenchymal stromal cells during bone repair. Stem Cell Reports 2022; 17:2318-2333. [PMID: 36150383 PMCID: PMC9561611 DOI: 10.1016/j.stemcr.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) transplantation could enhance bone repair. However, the cell fate of transplanted MSCs, in terms of their local distribution and spatial associations with other types of cells were poorly understood. Here, we developed a single-cell 3D spatial correlation (sc3DSC) method to track transplanted MSCs based on deep tissue microscopy of fluorescent nanoparticles (fNPs) and immunofluorescence of key proteins. Locally delivered fNP-labeled MSCs enhanced tibial defect repair, increased the number of stem cells and vascular maturity in mice. fNP-MSCs persisted in the defect throughout repair. But only a small portion of transplanted cells underwent osteogenic differentiation (OSX+); a significant portion has maintained their expression of mesenchymal stem cell and skeletal stem cell markers (SCA-1 and PRRX1). Our results contribute to the optimization of MSC-based therapies. The sc3DSC method may be useful in studying cell-based therapies for the regeneration of other tissue types or disease models. Transplanted marrow stromal cells associated with vessels during bone defect repair Small proportion of transplanted cells differentiated into osteogenic cells A proportion of transplanted cells maintained expressions of stem cell markers
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zeshun Li
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Runpeng Hou
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Linhao Fu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Decheng Wu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Quanying Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kai Li
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
19
|
Jouan Y, Bouchemla Z, Bardèche-Trystram B, Sana J, Andrique C, Ea HK, Richette P, Latourte A, Cohen-Solal M, Hay E. Lin28a induces SOX9 and chondrocyte reprogramming via HMGA2 and blunts cartilage loss in mice. SCIENCE ADVANCES 2022; 8:eabn3106. [PMID: 36026443 PMCID: PMC9417174 DOI: 10.1126/sciadv.abn3106] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
Articular cartilage has low regenerative capacity despite permanent stress. Irreversible cartilage lesions characterize osteoarthritis (OA); this is not followed by tissue repair. Lin28a, an RNA binding protein, is detected in damaged cartilage in humans and mice. We investigated the role of LIN28a in cartilage physiology and in osteoarthritis. Lin28a-inducible conditional cartilage deletion up-regulated Mmp13 in intact mice and exacerbated the cartilage destruction in OA mice. Lin28a-specific cartilage overexpression protected mice against cartilage breakdown, stimulated chondrocyte proliferation and the expression of Prg4 and Sox9, and down-regulated Mmp13. Lin28a overexpression inhibited Let-7b and Let-7c miRNA levels while RNA-sequencing analysis revealed five genes of transcriptional factors regulated by Let-7. Moreover, Lin28a overexpression up-regulated HMGA2 and activated SOX9 transcription, a factor required for chondrocyte reprogramming. HMGA2 siRNA knockdown inhibited the cartilage protective effect of Lin28a overexpression. This study provides insights into a new pathway including the Lin28a-Let7 axis, thus promoting chondrocyte anabolism in injured cartilage in mice.
Collapse
Affiliation(s)
- Yohan Jouan
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Zohra Bouchemla
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | | | - Joanna Sana
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Caroline Andrique
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Hang-Korng Ea
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
- Hôpital Lariboisière, APHP, Paris, France
| | - Pascal Richette
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
- Hôpital Lariboisière, APHP, Paris, France
| | - Augustin Latourte
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| | - Martine Cohen-Solal
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
- Hôpital Lariboisière, APHP, Paris, France
| | - Eric Hay
- Bioscar UMR Inserm 1132 and Université de Paris, F-75010 Paris, France
| |
Collapse
|
20
|
Zhang J, Tong Y, Liu Y, Lin M, Xiao Y, Liu C. Mechanical loading attenuated negative effects of nucleotide analogue reverse-transcriptase inhibitor TDF on bone repair via Wnt/β-catenin pathway. Bone 2022; 161:116449. [PMID: 35605959 DOI: 10.1016/j.bone.2022.116449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/19/2022]
Abstract
The nucleotide analog reverse-transcriptase inhibitor, tenofovir disoproxil fumarate (TDF), is widely used to treat hepatitis B virus (HBV) and human immunodeficiency virus infection (HIV). However, long-term TDF usage is associated with an increased incidence of bone loss, osteoporosis, fractures, and other adverse reactions. We investigated the effect of chronic TDF use on bone homeostasis and defect repair in mice. In vitro, TDF inhibited osteogenic differentiation and mineralization in MC3T3-E1 cells. In vivo, 8-week-old C57BL/6 female mice were treated with TDF for 38 days to simulate chronic medication. Four-point bending test and μCT showed reduced bone biomechanical properties and microarchitecture in long bones. To investigate the effects of TDF on bone defect repair, we utilized a bilateral tibial monocortical defect model. μCT showed that TDF reduced new bone mineral tissue and bone mineral density (BMD) in the defect. To verify whether mechanical stimulation may be a useful treatment to counteract the negative bone effects of TDF, controlled dynamic mechanical loading was applied to the whole tibia during the matrix deposition phase on post-surgery days (PSDs) 5 to 8. Second harmonic generation (SHG) of collagen fibers and μCT showed that the reduction of new bone volume and bone mineral density caused by TDF was reversed by mechanical loading in the defect. Immunofluorescent deep tissue imaging showed that chronic TDF treatment reduced the number of osteogenic cells and the volume of new vessels. In addition, chronic TDF treatment inhibited the expressions of periostin and β-catenin, but increased the expression of sclerostin. Both negative effects were reversed by mechanical loading. Our study provides strong evidence that chronic use of TDF exerts direct and inhibitory impacts on bone repair, but appropriate mechanical loading could reverse these adverse effects.
Collapse
Affiliation(s)
- Jianing Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Yanrong Tong
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Yang Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Minmin Lin
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Yao Xiao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
21
|
Paul GR, Vallaster P, Rüegg M, Scheuren AC, Tourolle DC, Kuhn GA, Wehrle E, Müller R. Tissue-Level Regeneration and Remodeling Dynamics are Driven by Mechanical Stimuli in the Microenvironment in a Post-Bridging Loaded Femur Defect Healing Model in Mice. Front Cell Dev Biol 2022; 10:856204. [PMID: 35686050 PMCID: PMC9171432 DOI: 10.3389/fcell.2022.856204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/07/2022] [Indexed: 11/30/2022] Open
Abstract
Bone healing and remodeling are mechanically driven processes. While the generalized response to mechanical stimulation in bone is well-understood, much less is known about the mechanobiology-regulating tissue-scale bone formation and resorption during the reparative and remodeling phases of fracture healing. In this study, we combined computational approaches in the form of finite element analysis and experimental approaches by using a loaded femoral defect model in mice to investigate the role of mechanical stimulation in the microenvironment of bone. Specifically, we used longitudinal micro-computed tomography to observe temporal changes in bone at different densities and micro-finite element analysis to map the mechanics of the microenvironment to tissue-scale formation, quiescence (no change in bone presence between time points), and resorption dynamics in the late reparative and remodeling phases (post bridging). Increasing levels of effective strain led to increasing conditional probability of bone formation, while decreasing levels of effective strain led to increasing probability of bone resorption. In addition, the analysis of mineralization dynamics showed both a temporal and effective strain level-dependent behavior. A logarithmic-like response was displayed, where the conditional probability of bone formation or resorption increased rapidly and plateaued or fell rapidly and plateaued as mechanical strain increased.
Collapse
|
22
|
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
|
23
|
Buettmann EG, Yoneda S, Hu P, McKenzie JA, Silva MJ. Postnatal Osterix but not DMP1 lineage cells significantly contribute to intramembranous ossification in three preclinical models of bone injury. Front Physiol 2022; 13:1083301. [PMID: 36685200 PMCID: PMC9846510 DOI: 10.3389/fphys.2022.1083301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/02/2022] [Indexed: 01/06/2023] Open
Abstract
Murine models of long-bone fracture, stress fracture, and cortical defect are used to discern the cellular and molecular mediators of intramembranous and endochondral bone healing. Previous work has shown that Osterix (Osx+) and Dentin Matrix Protein-1 (DMP1+) lineage cells and their progeny contribute to injury-induced woven bone formation during femoral fracture, ulnar stress fracture, and tibial cortical defect repair. However, the contribution of pre-existing versus newly-derived Osx+ and DMP1+ lineage cells in these murine models of bone injury is unclear. We addressed this knowledge gap by using male and female 12-week-old, tamoxifen-inducible Osx Cre_ERT2 and DMP1 Cre_ERT2 mice harboring the Ai9 TdTomato reporter allele. To trace pre-existing Osx+ and DMP1+ lineage cells, tamoxifen (TMX: 100 mg/kg gavage) was given in a pulse manner (three doses, 4 weeks before injury), while to label pre-existing and newly-derived lineage Osx+ and DMP1+ cells, TMX was first given 2 weeks before injury and continuously (twice weekly) throughout healing. TdTomato positive (TdT+) cell area and cell fraction were quantified from frozen histological sections of injured and uninjured contralateral samples at times corresponding with active woven bone formation in each model. We found that in uninjured cortical bone tissue, Osx Cre_ERT2 was more efficient than DMP1 Cre_ERT2 at labeling the periosteal and endosteal surfaces, as well as intracortical osteocytes. Pulse-labeling revealed that pre-existing Osx+ lineage and their progeny, but not pre-existing DMP1+ lineage cells and their progeny, significantly contributed to woven bone formation in all three injury models. In particular, these pre-existing Osx+ lineage cells mainly lined new woven bone surfaces and became embedded as osteocytes. In contrast, with continuous dosing, both Osx+ and DMP1+ lineage cells and their progeny contributed to intramembranous woven bone formation, with higher TdT+ tissue area and cell fraction in Osx+ lineage versus DMP1+ lineage calluses (femoral fracture and ulnar stress fracture). Similarly, Osx+ and DMP1+ lineage cells and their progeny significantly contributed to endochondral callus regions with continuous dosing only, with higher TdT+ chondrocyte fraction in Osx+ versus DMP1+ cell lineages. In summary, pre-existing Osx+ but not DMP1+ lineage cells and their progeny make up a significant amount of woven bone cells (particularly osteocytes) across three preclinical models of bone injury. Therefore, Osx+ cell lineage modulation may prove to be an effective therapy to enhance bone regeneration.
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States.,Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Pei Hu
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
24
|
Individualized cyclic mechanical loading improves callus properties during the remodelling phase of fracture healing in mice as assessed from time-lapsed in vivo imaging. Sci Rep 2021; 11:23037. [PMID: 34845246 PMCID: PMC8630002 DOI: 10.1038/s41598-021-02368-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/12/2021] [Indexed: 01/15/2023] Open
Abstract
Fracture healing is regulated by mechanical loading. Understanding the underlying mechanisms during the different healing phases is required for targeted mechanical intervention therapies. Here, the influence of individualized cyclic mechanical loading on the remodelling phase of fracture healing was assessed in a non-critical-sized mouse femur defect model. After bridging of the defect, a loading group (n = 10) received individualized cyclic mechanical loading (8–16 N, 10 Hz, 5 min, 3 × /week) based on computed strain distribution in the mineralized callus using animal-specific real-time micro-finite element analysis with 2D/3D visualizations and strain histograms. Controls (n = 10) received 0 N treatment at the same post-operative time-points. By registration of consecutive scans, structural and dynamic callus morphometric parameters were followed in three callus sub-volumes and the adjacent cortex showing that the remodelling phase of fracture healing is highly responsive to cyclic mechanical loading with changes in dynamic parameters leading to significantly larger formation of mineralized callus and higher degree of mineralization. Loading-mediated maintenance of callus remodelling was associated with distinct effects on Wnt-signalling-associated molecular targets Sclerostin and RANKL in callus sub-regions and the adjacent cortex (n = 1/group). Given these distinct local protein expression patterns induced by cyclic mechanical loading during callus remodelling, the femur defect loading model with individualized load application seems suitable to further understand the local spatio-temporal mechano-molecular regulation of the different fracture healing phases.
Collapse
|
25
|
Wang Z, Liu Y, Zhang J, Lin M, Xiao C, Bai H, Liu C. Mechanical loading alleviated the inhibition of β2-adrenergic receptor agonist terbutaline on bone regeneration. FASEB J 2021; 35:e22033. [PMID: 34739146 DOI: 10.1096/fj.202101045rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022]
Abstract
The long-term use of adrenergic medication in treating various conditions, such as asthma, increases the chances of bone fracture. Dynamic mechanical loading at a specific time is a method for improving bone quality and promoting healing. Therefore, we hypothesized that precisely controlling the mechanical environment can contribute to the alleviation of the negative effects of chronic treatment with the common asthma drug terbutaline, which is a β2-adrenergic receptor agonist that facilitates bone homeostasis and defect repair through its anabolic effect on osteogenic cells. Our in vitro results showed that terbutaline can directly inhibit osteogenesis by impairing osteogenic differentiation and mineralization. Chronic treatment in vivo was simulated by administering terbutaline to C57BL/6J mice for 4 weeks before bone defect surgery and mechanical loading. We utilized a stabilized tibial defect model, which allowed the application of anabolic mechanical loading. During homeostasis, chronic terbutaline treatment reduced the bone formation rate, the fracture toughness of long bones, and the concentrations of bone formation markers in the sera. During defect repair, terbutaline decreased the bone volume, type H vessel, and total blood vessel volume. Terbutaline treatment reduced the number of osteogenic cells. Periostin, which was secreted mainly by Prrx1+ osteoprogenitors and F4/80+ macrophages, was inhibited by treating the bone defect with terbutaline. Interestingly, controlled mechanical loading facilitated the recovery of bone volume and periostin expression and the number of osteogenic cells within the defect. In conclusion, mechanical loading can rescue negative effects on new bone accrual and repair induced by chronic terbutaline treatment.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jianing Zhang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chufan Xiao
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Haoying Bai
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
26
|
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
|
27
|
Borgiani E, Duda GN, Willie BM, Checa S. Bone morphogenetic protein 2-induced cellular chemotaxis drives tissue patterning during critical-sized bone defect healing: an in silico study. Biomech Model Mechanobiol 2021; 20:1627-1644. [PMID: 34047890 PMCID: PMC8298257 DOI: 10.1007/s10237-021-01466-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022]
Abstract
Critical-sized bone defects are critical healing conditions that, if left untreated, often lead to non-unions. To reduce the risk, critical-sized bone defects are often treated with recombinant human BMP-2. Although enhanced bone tissue formation is observed when BMP-2 is administered locally to the defect, spatial and temporal distribution of callus tissue often differs from that found during regular bone healing or in defects treated differently. How this altered tissue patterning due to BMP-2 treatment is linked to mechano-biological principles at the cellular scale remains largely unknown. In this study, the mechano-biological regulation of BMP-2-treated critical-sized bone defect healing was investigated using a multiphysics multiscale in silico approach. Finite element and agent-based modeling techniques were combined to simulate healing within a critical-sized bone defect (5 mm) in a rat femur. Computer model predictions were compared to in vivo microCT data outcome of bone tissue patterning at 2, 4, and 6 weeks postoperation. In vivo, BMP-2 treatment led to complete healing through periosteal bone bridging already after 2 weeks postoperation. Computer model simulations showed that the BMP-2 specific tissue patterning can be explained by the migration of mesenchymal stromal cells to regions with a specific concentration of BMP-2 (chemotaxis). This study shows how computational modeling can help us to further understand the mechanisms behind treatment effects on compromised healing conditions as well as to optimize future treatment strategies.
Collapse
Affiliation(s)
- Edoardo Borgiani
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Bettina M Willie
- Research Centre, Department of Pediatric Surgery, Shriners Hospital for Children-Canada, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| | - Sara Checa
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
28
|
Camal Ruggieri IN, Cícero AM, Issa JPM, Feldman S. Bone fracture healing: perspectives according to molecular basis. J Bone Miner Metab 2021; 39:311-331. [PMID: 33151416 DOI: 10.1007/s00774-020-01168-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
Fractures have a great impact on health all around the world and with fracture healing optimization; this problem could be resolved partially. To make a practical contribution to this issue, the knowledge of bone tissue, cellularity, and metabolism is essential, especially cytoskeletal architecture and its transformations according to external pressures. Special physical and chemical characteristics of the extracellular matrix (ECM) allow the transmission of mechanical stimuli from outside the cell to the plasmatic membrane. The osteocyte cytoskeleton is conformed by a complex network of actin and microtubules combined with crosslinker proteins like vinculin and fimbrin, connecting and transmitting outside stimuli through EMC to cytoplasm. Herein, critical signaling pathways like Cx43-depending ones, MAPK/ERK, Wnt, YAP/TAZ, Rho-ROCK, and others are activated due to mechanical stimuli, resulting in osteocyte cytoskeletal changes and ECM remodeling, altering the tissue and, therefore, the bone. In recent years, the osteocyte has gained more interest and value in relation to bone homeostasis as a great coordinator of other cell populations, thanks to its unique functions. By integrating the latest advances in relation to intracellular signaling pathways, mechanotransmission system of the osteocyte and bone tissue engineering, there are promising experimental strategies, while some are ready for clinical trials. This work aims to show clearly and precisely the integration between cytoskeleton and main molecular pathways in relation to mechanotransmission mechanism in osteocytes, and the use of this theoretical knowledge in therapeutic tools for bone fracture healing.
Collapse
Affiliation(s)
- Iván Nadir Camal Ruggieri
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina.
| | - Andrés Mauricio Cícero
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
| | | | - Sara Feldman
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
- Research Council of the Rosario National University (CIUNR) and CONICET, Rosario, Argentina
| |
Collapse
|
29
|
Taguchi T, Lopez MJ. An overview of de novo bone generation in animal models. J Orthop Res 2021; 39:7-21. [PMID: 32910496 PMCID: PMC7820991 DOI: 10.1002/jor.24852] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 02/04/2023]
Abstract
Some of the earliest success in de novo tissue generation was in bone tissue, and advances, facilitated by the use of endogenous and exogenous progenitor cells, continue unabated. The concept of one health promotes shared discoveries among medical disciplines to overcome health challenges that afflict numerous species. Carefully selected animal models are vital to development and translation of targeted therapies that improve the health and well-being of humans and animals alike. While inherent differences among species limit direct translation of scientific knowledge between them, rapid progress in ex vivo and in vivo de novo tissue generation is propelling revolutionary innovation to reality among all musculoskeletal specialties. This review contains a comparison of bone deposition among species and descriptions of animal models of bone restoration designed to replicate a multitude of bone injuries and pathology, including impaired osteogenic capacity.
Collapse
Affiliation(s)
- Takashi Taguchi
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary MedicineLouisiana State UniversityBaton RougeLouisianaUSA
| | - Mandi J. Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary MedicineLouisiana State UniversityBaton RougeLouisianaUSA
| |
Collapse
|
30
|
Plecko M, Klein K, Planzer K, Wähnert D, Behm P, Ferguson SJ, Brianza S, Stadelmann VA, von Rechenberg B. Variable fixation promotes callus formation: an experimental study on transverse tibial osteotomies stabilized with locking plates. BMC Musculoskelet Disord 2020; 21:806. [PMID: 33272239 PMCID: PMC7713143 DOI: 10.1186/s12891-020-03781-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/09/2020] [Indexed: 12/31/2022] Open
Abstract
Background A new locking screw technology, named variable fixation, has been developed aiming at promoting bone callus formation providing initial rigid fixation followed by progressive fracture gap dynamisation. In this study, we compared bone callus formation in osteotomies stabilized with standard locking fixation against that of osteotomies stabilized with variable fixation in an established tibia ovine model. Methods A 3 mm tibial transverse osteotomy gap was stabilized in three groups of six female sheep each with a locking plate and either 1) standard fixation in both segments (group LS) or 2) variable fixation in the proximal and standard fixation in the distal bone segment (group VFLS3) or 3) variable fixation in both segments (group VFLS6). The implantation site and fracture healing were compared between groups by means of radiologic, micro tomographic, biomechanical, and histological investigations. Results Compared to LS callus, VFLS3 callus was 40% larger and about 3% denser, while VFLS6 callus was 93% larger and its density about 7.2% lower. VFLS3 showed 65% and VFLS6 163% larger amount of callus at the cis-cortex. There wasn’t a significant difference in the amount of callus at the cis and trans-cortex in groups featuring variable fixation only. Investigated biomechanical variables were not significantly different among groups and histology showed comparable good healing in all groups. Tissues adjacent to the implants did not show any alteration of the normal structure in all groups. Conclusions Variable fixation promoted the formation of a larger amount of bone callus, equally distributed at the cis and trans cortices. The histological and biomechanical properties of the variable fixation callus were equivalent to those of the standard fixation callus. The magnitude of variable fixation had a biological effect on the formation of bone callus. At the implantation site, the usage of variable fixation did not raise additional concerns with respect to standard fixation. The formation of a larger amount of mature callus suggests that fractures treated with variable fixation might have a higher probability to bridge the fracture gap. The conditions where its usage can be most beneficial for patients needs to be clinically defined.
Collapse
Affiliation(s)
- Michael Plecko
- Trauma Hospital Graz (UKH), Goestinger Strasse 24, 8021, Graz, Austria
| | - Karina Klein
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Katrin Planzer
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Dirk Wähnert
- Protestant Hospital of Bethel Foundation, Department of Trauma and Orthopedic Surgery, Bielefeld, Germany
| | - Pascal Behm
- Institute for Biomechanics, ETH, Zurich, Switzerland
| | - Stephen J Ferguson
- Institute for Biomechanics, ETH, Zurich, Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Stefano Brianza
- Biomech Innovations AG, Aarbergstrasse 5, CH-2560, Nidau, Switzerland.
| | | | - Brigitte von Rechenberg
- Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Frank A, Brianza S, Plecko M, Raschke MJ, Wähnert D. Variable Fixation Technology Provides Rigid as Well as Progressive Dynamic Fixation: A Biomechanical Investigation. J Bone Joint Surg Am 2020; 102:e115. [PMID: 33086351 DOI: 10.2106/jbjs.19.01302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND A new locking-screw technology, the Variable Fixation Locking Screw (VFLS; Biomech Innovations), was developed with the aim of promoting secondary fracture-healing. The VFLS features a resorbable sleeve that progressively decreases its mechanical properties and mass during the fracture-healing time. In this study, we investigated whether the VFLS can provide rigid as well as progressive dynamic fixation. METHODS The interfragmentary stability provided by the VFLS was tested in a simulated fracture-gap model and compared with that provided by standard locking or by a combination of both technologies under compression and torsional loading. Tests were performed with an intact sleeve (initial condition) and after its chemical dissolution. An optical measurement system was used to characterize interfragmentary movements. RESULTS The axial stiffness did not differ significantly among groups in the initial condition. Sleeve resorption significantly decreased construct stiffness. The torsional stiffness of the samples instrumented with the VFLS was lower than that of the control group. The degradation of the sleeve resulted in a significant increase in axial displacement recorded at both the cis and trans cortices. In samples featuring combined technologies, this increase was about 12% to 20% at the trans cortex and about 50% to 60% at the cis cortex. In samples featuring VFLS technology only, this increase was about 20% to 37% at the trans cortex and about 70% to 125% at the cis cortex. CONCLUSIONS The initial stability offered by the VFLS is equivalent to that of standard locking-screw technology. The resorption of the degradable sleeve leads to effective and reproducible fracture-gap dynamization, progressively varying the way the fracture gap is strained and the magnitude of the strain. CLINICAL RELEVANCE The VFLS provides rigid and progressive dynamic fixation in vitro. Such variable stability might have beneficial effects in terms of triggering and boosting secondary fracture-healing.
Collapse
Affiliation(s)
- A Frank
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
- Institute for Musculoskeletal Medicine, Westfaelische-Wilhelms-University Muenster, Muenster, Germany
| | - S Brianza
- Biomech Innovations AG, Nidau, Switzerland
| | - M Plecko
- Unfallkrankenhaus Steiermark, Graz, Austria
| | - M J Raschke
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - D Wähnert
- Institute for Musculoskeletal Medicine, Westfaelische-Wilhelms-University Muenster, Muenster, Germany
- Department of Orthopedic and Trauma Surgery, Protestant Hospital of Bethel Foundation, Bielefeld, Germany
| |
Collapse
|
32
|
Mu Z, Chen K, Yuan S, Li Y, Huang Y, Wang C, Zhang Y, Liu W, Luo W, Liang P, Li X, Song J, Ji P, Cheng F, Wang H, Chen T. Gelatin Nanoparticle-Injectable Platelet-Rich Fibrin Double Network Hydrogels with Local Adaptability and Bioactivity for Enhanced Osteogenesis. Adv Healthc Mater 2020; 9:e1901469. [PMID: 31994326 DOI: 10.1002/adhm.201901469] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/19/2019] [Indexed: 12/11/2022]
Abstract
Bone healing is a dynamic process regulated by biochemical signals such as chemokines and growth factors, and biophysical signals such as topographical and mechanical features of extracellular matrix or mechanical stimuli. Hereby, a mechanically tough and bioactive hydrogel based on autologous injectable platelet-rich fibrin (iPRF) modified with gelatin nanoparticles (GNPs) is developed. This composite hydrogel demonstrates a double network (DN) mechanism, wherein covalent network of fibrin serves to maintain material integrity, and self-assembled colloidal network of GNPs dissipates force upon loading. A rabbit sinus augmentation model is used to investigate the bioactivity and osteogenesis capacity of the DN hydrogels. The DN hydrogels adapt to the local environmental complexity of bone defects, i.e., accommodate the irregular shape of the defects and withstand the pressure formed in the maxillary sinus during animal's respiration process. The DN hydrogel is also demonstrated to absorb and prolong the release of the bioactive growth factors stemming from iPRF, which could have contributed to the early angiogenesis and osteogenesis observed inside the sinus. This adaptable and bioactive DN hydrogel can achieve enhanced bone regeneration in treating complex bone defects by maintaining long-term bone mass and withstanding the functional mechanical stimuli.
Collapse
Affiliation(s)
- Zhixiang Mu
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Kaiwen Chen
- Key State Laboratory of Fine ChemicalsSchool of BioengineeringDalian University of Technology No. 2 Linggong Road, High‐tech District Dalian 116024 P. R. China
| | - Shuai Yuan
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Yihan Li
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Yuanding Huang
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Chao Wang
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Yang Zhang
- Laboratory of Regenerative BiomaterialsDepartment of Biomedical EngineeringHealth Science CenterShenzhen University Shenzhen Guangdong Province 518037 P. R. China
| | - Wenzhao Liu
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Wenping Luo
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Panpan Liang
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Xiaodong Li
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Jinlin Song
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Ping Ji
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| | - Fang Cheng
- Key State Laboratory of Fine ChemicalsSchool of Chemical EngineeringDalian University of Technology No. 2 Linggong Road, High‐tech District Dalian 116024 P. R. China
| | - Huanan Wang
- Key State Laboratory of Fine ChemicalsSchool of BioengineeringDalian University of Technology No. 2 Linggong Road, High‐tech District Dalian 116024 P. R. China
| | - Tao Chen
- Laboratory of Oral Diseases and Biomedical SciencesChongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqing Medical University Chongqing 401147 P. R. China
| |
Collapse
|
33
|
Comparative effectiveness of PEEK rods versus titanium alloy rods in cervical fusion in a new sheep model. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2020; 29:1159-1166. [PMID: 32008100 DOI: 10.1007/s00586-020-06307-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 01/10/2020] [Accepted: 01/18/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pedicle screw and rod instrumentation based on titanium can produce satisfying strength and stiffness for spinal fusion. However, excessive stiffness produced by titanium rods may cause stress shielding. Thus, polyetheretherketone (PEEK) rods with a low modulus of elasticity were introduced as substitutes for titanium rods. The purpose of this paper is to compare the effectiveness of PEEK rods versus titanium alloy rods in anterior spinal fusion with a new sheep model. METHODS Sheep models of anterior-posterior cervical fusion were innovatively adopted in our study. Twenty-four sheep were randomly divided into a control group and a treatment group that received anterior-posterior cervical fixation with titanium rods or PEEK rods, respectively. Then, surgical segments were harvested and assessed by X-ray, micro-CT and histological examination to evaluate the efficiency of bone fusion. RESULTS No complications related to fixation were found during the research process. The results of the X-ray showed a stronger spinal fusion in the PEEK rod groups than in the titanium rod group at 12 weeks postoperatively, and both groups underwent bone fusion at 24 weeks postoperatively. The results of micro-CT showed that fixation with PEEK rods achieved better bone ingrowth at an early postoperative stage (12 weeks) compared to fixation with titanium rods (bone volume fraction (BVF): 20.26 ± 4.36% vs 14.48 ± 3.49%, p < 0.05). The same trend was detected in the histological analysis, where the mineralized bone fraction in the experiment group (21.01 ± 3.48%) was significantly higher than that in the control group (16.73 ± 2.95%). In addition, better osseointegration was found in the experiment group at the early postoperative stage at 12 weeks (bone apposition (BA): 16.22 ± 3.24% vs 11.67 ± 3.63%, p < 0.05). However, there were no significant differences at 24 weeks postoperatively. CONCLUSION PEEK rods can be used safely in a sheep model of anterior-posterior cervical fixation. Compared to traditional titanium rods, earlier and more evident bone fusion was found in the PEEK rods group. These slides can be retrieved under Electronic Supplementary Material.
Collapse
|
34
|
Alcântara ACS, Assis I, Prada D, Mehle K, Schwan S, Costa-Paiva L, Skaf MS, Wrobel LC, Sollero P. Patient-Specific Bone Multiscale Modelling, Fracture Simulation and Risk Analysis-A Survey. MATERIALS (BASEL, SWITZERLAND) 2019; 13:E106. [PMID: 31878356 PMCID: PMC6981613 DOI: 10.3390/ma13010106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/26/2022]
Abstract
This paper provides a starting point for researchers and practitioners from biology, medicine, physics and engineering who can benefit from an up-to-date literature survey on patient-specific bone fracture modelling, simulation and risk analysis. This survey hints at a framework for devising realistic patient-specific bone fracture simulations. This paper has 18 sections: Section 1 presents the main interested parties; Section 2 explains the organzation of the text; Section 3 motivates further work on patient-specific bone fracture simulation; Section 4 motivates this survey; Section 5 concerns the collection of bibliographical references; Section 6 motivates the physico-mathematical approach to bone fracture; Section 7 presents the modelling of bone as a continuum; Section 8 categorizes the surveyed literature into a continuum mechanics framework; Section 9 concerns the computational modelling of bone geometry; Section 10 concerns the estimation of bone mechanical properties; Section 11 concerns the selection of boundary conditions representative of bone trauma; Section 12 concerns bone fracture simulation; Section 13 presents the multiscale structure of bone; Section 14 concerns the multiscale mathematical modelling of bone; Section 15 concerns the experimental validation of bone fracture simulations; Section 16 concerns bone fracture risk assessment. Lastly, glossaries for symbols, acronyms, and physico-mathematical terms are provided.
Collapse
Affiliation(s)
- Amadeus C. S. Alcântara
- Department of Computational Mechanics, School of Mechanical Engineering, University of Campinas—UNICAMP, Campinas, Sao Paulo 13083-860, Brazil; (A.C.S.A.); (D.P.)
| | - Israel Assis
- Department of Integrated Systems, School of Mechanical Engineering, University of Campinas—UNICAMP, Campinas, Sao Paulo 13083-860, Brazil;
| | - Daniel Prada
- Department of Computational Mechanics, School of Mechanical Engineering, University of Campinas—UNICAMP, Campinas, Sao Paulo 13083-860, Brazil; (A.C.S.A.); (D.P.)
| | - Konrad Mehle
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, 06217 Merseburg, Germany;
| | - Stefan Schwan
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, 06120 Halle/Saale, Germany;
| | - Lúcia Costa-Paiva
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas—UNICAMP, Campinas, Sao Paulo 13083-887, Brazil;
| | - Munir S. Skaf
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas—UNICAMP, Campinas, Sao Paulo 13083-860, Brazil;
| | - Luiz C. Wrobel
- Institute of Materials and Manufacturing, Brunel University London, Uxbridge UB8 3PH, UK;
- Department of Civil and Environmental Engineering, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro 22451-900, Brazil
| | - Paulo Sollero
- Department of Computational Mechanics, School of Mechanical Engineering, University of Campinas—UNICAMP, Campinas, Sao Paulo 13083-860, Brazil; (A.C.S.A.); (D.P.)
| |
Collapse
|
35
|
Strain Distribution Evaluation of Rat Tibia under Axial Compressive Load by Combining Strain Gauge Measurement and Finite Element Analysis. Appl Bionics Biomech 2019; 2019:1736763. [PMID: 31871486 PMCID: PMC6913262 DOI: 10.1155/2019/1736763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/16/2019] [Accepted: 11/12/2019] [Indexed: 11/18/2022] Open
Abstract
This study is aimed at providing an effective method for determining strain-load relationship and at quantifying the strain distribution within the whole tibia under axial compressive load on rats. Rat tibial models with axial compressive load were designed. Strains in three directions (0°, 45°, and 90°) at the proximal shaft of the tibia were measured by using a strain gauge rosette, which was used to calculate the maximum and minimum principal strains. Moreover, the strain at the midshaft of the tibia was measured by a single-element strain gauge. The slopes of the strain-load curves with different peak loads were calculated to assess the stability of the strain gauge measurement. Mechanical environment in the whole tibia by the axial compressive load was quantified using finite element analysis (FEA) based on microcomputed tomography images. The von Mises elastic strain distributions of the whole tibiae were evaluated. Slopes of the strain-load curves showed no significant differences among different peak loads (ANOVA; P > 0.05), indicating that the strain-load relationship obtained from the strain gauge measurement was reasonable and stable. The FEA results corresponded to the experimental results with an error smaller than 15% (paired Student's t-test, P > 0.05), signifying that the FEA can simulate the experiment reasonably. FEA results showed that the von Mises elastic strain was the lowest in the middle and gradually increased to both sides along the lateral direction, with the maximal von Mises elastic strain being observed on the posterior side under the distal tibiofibular synostosis. The method of strain gauge measurements and FEA used in this study can provide a feasible way to obtain the mechanical environment of the tibiae under axial compressive load on the rats and serve as a reference for further exploring the mechanical response of the bone by axial compressive load.
Collapse
|
36
|
Delgado-Ruiz RA, Calvo-Guirado JL, Romanos GE. Effects of occlusal forces on the peri-implant-bone interface stability. Periodontol 2000 2019; 81:179-193. [PMID: 31407438 DOI: 10.1111/prd.12291] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The occlusal forces and their influence on the initiation of peri-implant bone loss or their relationship with peri-implantitis have created discussion during the past 30 years given the discrepancies observed in clinical, animal, and finite element analysis studies. Beyond these contradictions, in the case of an osseointegrated implant, the occlusal forces can influence the implant-bone interface and the cells responsible for the bone remodeling in different ways that may result in the maintenance or loss of the osseointegration. This comprehensive review focuses on the information available about the forces transmitted through the implant-crown system to the implant-bone interface and the mechano-transduction phenomena responsible for the bone cells' behavior and their interactions. Knowledge of the basic molecular biology of the peri-implant bone would help clinicians to understand the complex phenomenon of occlusal forces and their effects on the implant-bone interface, and would allow better control of the negative effects of mechanical stresses, leading to therapy with fewer risks and complications.
Collapse
Affiliation(s)
- Rafael Arcesio Delgado-Ruiz
- Department of Prosthodontics and Digital Technology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Jose Luis Calvo-Guirado
- International Dentistry Research Cathedra, Faculty of Medicine and Dentistry, Universidad Catolica San Antonio De Murcia (UCAM), Murcia, Spain
| | - Georgios E Romanos
- Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA.,Department of Oral Surgery and Implant Dentistry, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|
37
|
Costa Fernandes CJD, Zambuzzi WF. Fibroblast-secreted trophic factors contribute with ECM remodeling stimulus and upmodulate osteocyte gene markers in osteoblasts. Biochimie 2019; 168:92-99. [PMID: 31676316 DOI: 10.1016/j.biochi.2019.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022]
Abstract
As osteogenesis is a multifactorial mechanism, we wonder whether osteoblast-induced extracellular matrix (ECM) remodeling might be modulated by trophic factors released by fibroblasts in a paracrine signaling manner. To address this issue, fibroblasts were cultured for 72 h under conventional conditions when their conditioned medium was harvested and used to challenge pre-osteoblasts (MC3T3-E1 cells) for 14 days. Preliminarily, we validated the potential effect of fibroblasts in contributing to osteocyte phenotype, which specifically requires significant expression of Dentin Matrix Protein 1 (DMP1; about 10-fold changes) and Sclerostin (SOST; about 7-fold changes), both biomarkers of osteocyte. Fibroblasts also seem contributing to ECM remodeling in osteoblasts, because we detected a high level of both mRNA and enzyme activities of matrix metalloproteinase -9 (MMP-9) as well as a high level of reversion inducing cysteine rich protein with kazal motifs (RECK) transcripts (about 13-fold changes), a membrane-anchored MMP inhibitor, which seems to be a constitutive pathway in osteoblasts. Considering inflammatory panorama and using RTqPCR technology, both IL-13 (about 13-fold changes) and IL-33 (about 5-fold changes) genes were up-expressed in response to the fibroblast-secreted trophic factors, as were the receptor activator of NF-κB ligand (RANKL; about 8-fold changes) and osteoprotegerin (OPG; about 3-fold changes). Although preliminary, these data suggest a stimulus to finely control osteoclastogenesis, and this mechanism reinforces the role of fibroblasts in bone remodeling and homeostasis. Moreover, these results suggest an important crosstalk between fibroblast and osteoblast, when fibroblast-secreted trophic factors upmodulate osteocyte gene markers and contribute to ECM remodeling stimulus in osteoblast.
Collapse
Affiliation(s)
- Célio Jr da Costa Fernandes
- Lab. of Bioassays and Cell Dynamics, Department of Chemistry and Biochemistry, Institute of Biosciences, Universidade Estadual Paulista, UNESP, CEP 18618-970, Botucatu, São Paulo, Brazil
| | - Willian Fernando Zambuzzi
- Lab. of Bioassays and Cell Dynamics, Department of Chemistry and Biochemistry, Institute of Biosciences, Universidade Estadual Paulista, UNESP, CEP 18618-970, Botucatu, São Paulo, Brazil.
| |
Collapse
|
38
|
Buettmann EG, McKenzie JA, Migotsky N, Sykes DA, Hu P, Yoneda S, Silva MJ. VEGFA From Early Osteoblast Lineage Cells (Osterix+) Is Required in Mice for Fracture Healing. J Bone Miner Res 2019; 34:1690-1706. [PMID: 31081125 PMCID: PMC6744295 DOI: 10.1002/jbmr.3755] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/23/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
Bone formation via intramembranous and endochondral ossification is necessary for successful healing after a wide range of bone injuries. The pleiotropic cytokine, vascular endothelial growth factor A (VEGFA) has been shown, via nonspecific pharmacologic inhibition, to be indispensable for angiogenesis and ossification following bone fracture and cortical defect repair. However, the importance of VEGFA expression by different cell types during bone healing is not well understood. We sought to determine the role of VEGFA from different osteoblast cell subsets following clinically relevant models of bone fracture and cortical defect. Ubiquitin C (UBC), Osterix (Osx), or Dentin matrix protein 1 (Dmp1) Cre-ERT2 mice (male and female) containing floxed VEGFA alleles (VEGFAfl/fl ) were either given a femur full fracture, ulna stress fracture, or tibia cortical defect at 12 weeks of age. All mice received tamoxifen continuously starting 2 weeks before bone injury and throughout healing. UBC Cre-ERT2 VEGFAfl/fl (UBC cKO) mice, which were used to mimic nonspecific inhibition, had minimal bone formation and impaired angiogenesis across all bone injury models. UBC cKO mice also exhibited impaired periosteal cell proliferation during full fracture, but not stress fracture repair. Osx Cre-ERT2 VEGFAfl/fl (Osx cKO) mice, but not Dmp1 Cre-ERT2 VEGFAfl/fl (Dmp1 cKO) mice, showed impaired periosteal bone formation and angiogenesis in models of full fracture and stress fracture. Neither Osx cKO nor Dmp1 cKO mice demonstrated significant impairments in intramedullary bone formation and angiogenesis following cortical defect. These data suggest that VEGFA from early osteolineage cells (Osx+), but not mature osteoblasts/osteocytes (Dmp1+), is critical at the time of bone injury for rapid periosteal angiogenesis and woven bone formation during fracture repair. Whereas VEGFA from another cell source, not from the osteoblast cell lineage, is necessary at the time of injury for maximum cortical defect intramedullary angiogenesis and osteogenesis. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicole Migotsky
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - David Aw Sykes
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Pei Hu
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
39
|
Cabahug-Zuckerman P, Liu C, Cai C, Mahaffey I, Norman SC, Cole W, Castillo AB. Site-Specific Load-Induced Expansion of Sca-1 +Prrx1 + and Sca-1 -Prrx1 + Cells in Adult Mouse Long Bone Is Attenuated With Age. JBMR Plus 2019; 3:e10199. [PMID: 31667455 PMCID: PMC6808224 DOI: 10.1002/jbm4.10199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 02/04/2023] Open
Abstract
Aging is associated with significant bone loss and increased fracture risk, which has been attributed to a diminished response to anabolic mechanical loading. In adults, skeletal progenitors proliferate and differentiate into bone‐forming osteoblasts in response to increasing mechanical stimuli, though the effects of aging on this response are not well‐understood. Here we show that both adult and aged mice exhibit load‐induced periosteal bone formation, though the response is significantly attenuated with age. We also show that the acute response of adult bone to loading involves expansion of Sca‐1+Prrx1+ and Sca‐1−Prrx1+ cells in the periosteum. On the endosteal surface, loading enhances proliferation of both these cell populations, though the response is delayed by 2 days relative to the periosteal surface. In contrast to the periosteum and endosteum, the marrow does not exhibit increased proliferation of Sca‐1+Prrx1+ cells, but only of Sca‐1−Prrx1+ cells, underscoring fundamental differences in how the stem cell niche in distinct bone envelopes respond to mechanical stimuli. Notably, the proliferative response to loading is absent in aged bone even though there are similar baseline numbers of Prrx1 + cells in the periosteum and endosteum, suggesting that the proliferative capacity of progenitors is attenuated with age, and proliferation of the Sca‐1+Prrx1+ population is critical for load‐induced periosteal bone formation. These findings provide a basis for the development of novel therapeutics targeting these cell populations to enhance osteogenesis for overcoming age‐related bone loss. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Pamela Cabahug-Zuckerman
- Department of Orthopaedic Surgery NYU Langone Health, New York University New York NY USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University New York NY USA.,Rehabilitation Research and Development Veterans Affairs New York Harbor Healthcare System New York NY USA
| | - Chao Liu
- Department of Orthopaedic Surgery NYU Langone Health, New York University New York NY USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University New York NY USA.,Rehabilitation Research and Development Veterans Affairs New York Harbor Healthcare System New York NY USA
| | - Cinyee Cai
- Department of Orthopaedic Surgery NYU Langone Health, New York University New York NY USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University New York NY USA
| | - Ian Mahaffey
- Rehabilitation Research and Development Veterans Affairs Palo Alto Healthcare System Palo Alto CA USA
| | - Stephanie C Norman
- Rehabilitation Research and Development Veterans Affairs Palo Alto Healthcare System Palo Alto CA USA
| | - Whitney Cole
- Rehabilitation Research and Development Veterans Affairs Palo Alto Healthcare System Palo Alto CA USA
| | - Alesha B Castillo
- Department of Orthopaedic Surgery NYU Langone Health, New York University New York NY USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University New York NY USA.,Rehabilitation Research and Development Veterans Affairs New York Harbor Healthcare System New York NY USA
| |
Collapse
|
40
|
Liu C, Cabahug-Zuckerman P, Stubbs C, Pendola M, Cai C, Mann KA, Castillo AB. Mechanical Loading Promotes the Expansion of Primitive Osteoprogenitors and Organizes Matrix and Vascular Morphology in Long Bone Defects. J Bone Miner Res 2019; 34:896-910. [PMID: 30645780 PMCID: PMC8263903 DOI: 10.1002/jbmr.3668] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 12/17/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022]
Abstract
Elucidating the effects of mechanical stimulation on bone repair is crucial for optimization of the healing process. Specifically, the regulatory role that mechanical loading exerts on the osteogenic stem cell pool and vascular morphology during healing is incompletely understood. Because dynamic loading has been shown to enhance osteogenesis and repair, we hypothesized that loading induces the expansion of the osteoprogenitor cell population within a healing bone defect, leading to an increased presence of osteogenic cells. We further hypothesized that loading during the repair process regulates vascular and collagen matrix morphology and spatial interactions between vessels and osteogenic cells. To address these hypotheses, we used a mechanobiological bone repair model, which produces a consistent and reproducible intramembranous repair response confined in time and space. Bilateral tibial defects were created in adult C57BL/6 mice, which were subjected to axial compressive dynamic loading either during the early cellular invasion phase on postsurgical days (PSDs) 2 to 5 or during the matrix deposition phase on PSD 5 to 8. Confocal and two-photon microscopy was used to generate high-resolution three-dimensional (3D) renderings of longitudinal thick sections of the defect on PSD 10. Endomucin (EMCN)-positive vessels, Paired related homeobox 1 (Prrx1+) stem cell antigen-1 positive (Sca-1+) primitive osteoprogenitors (OPCs), and osterix positive (Osx+) preosteoblasts were visualized and quantified using deep tissue immunohistochemistry. New bone matrix was visualized with second harmonic generation autofluorescence of collagen fibers. We found that mechanical loading during the matrix deposition phase (PSD 5 to 8) increased vessel volume and number, and aligned vessels and collagen fibers to the load-bearing direction of bone. Furthermore, loading led to a significant increase in the proliferation and number of Prrx1+ Sca-1+ primitive OPCs, but not Osx+ preosteoblasts within the defect. Together, these data illustrate the adaptation of both collagen matrix and vascular morphology to better withstand mechanical load during bone repair, and that the mechanoresponsive cell population consists of the primitive osteogenic progenitors. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Chao Liu
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 10010
- Department of Orthopedic Surgery, School of Medicine, New York University, New York, NY 10010
- Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
| | - Pamela Cabahug-Zuckerman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 10010
- Department of Orthopedic Surgery, School of Medicine, New York University, New York, NY 10010
| | - Christopher Stubbs
- Department of Mechanical Engineering, New York University, New York, NY 10010
| | - Martin Pendola
- Department of Orthopedic Surgery, School of Medicine, New York University, New York, NY 10010
| | - Cinyee Cai
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 10010
| | - Kenneth A. Mann
- Department of Orthopedic Surgery, Upstate Medical University, New York, NY 13210
| | - Alesha B. Castillo
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 10010
- Department of Orthopedic Surgery, School of Medicine, New York University, New York, NY 10010
- Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
| |
Collapse
|
41
|
Exploring conditions that make cortical bone geometry optimal for physiological loading. Biomech Model Mechanobiol 2019; 18:1335-1349. [PMID: 30953214 DOI: 10.1007/s10237-019-01147-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/02/2019] [Indexed: 10/27/2022]
Abstract
While physiological loading on lower long bones changes during bone development, the bone cross section either remains circular or slowly changes from nearly circular to other shapes such as oval and roughly triangular. Bone is said to be an optimal structure, where strength is maximized using the optimal distribution of bone mass (also called Wolff's law). One of the most appropriate mathematical validations of this law would be a structural optimization-based formulation where total strain energy is minimized against a mass and a space constraint. Assuming that the change in cross section during bone development and homeostasis after adulthood is direct result of the change in physiological loading, this work investigates what optimization problem formulation (collectively, design variables, objective function, constraints, loading conditions, etc.) results in mathematically optimal solutions that resemble bones under actual physiological loading. For this purpose, an advanced structural optimization-based computational model for cortical bone development and defect repair is presented. In the optimization problem, overall bone stiffness is maximized first against a mass constraint, and then also against a polar first moment of area constraint that simultaneously constrains both mass and space. The investigation is completed in two stages. The first stage is developmental stage when physiological loading on lower long bones (tibia) is a random combination of axial, bending and torsion. The topology optimization applied to this case with the area moment constraint results into circular and elliptical cross sections similar to that found in growing mouse or human. The second investigation stage is bone homeostasis reached in adulthood when the physiological loading has a fixed pattern. A drill hole defect is applied to the adult mouse bone, which would disrupt the homeostasis. The optimization applied after the defect interestingly brings the damaged section back to the original intact geometry. The results, however, show that cortical bone geometry is optimal for the physiological loading only when there is also a constraint on polar moment of area. Further numerical experiments show that application of torsion along with the gait-analysis-based physiological loading improves the results, which seems to indicate that the cortical bone geometry is optimal for some amount of torsion in addition to the gait-based physiological loading. This work has a potential to be extended to bone growth/development models and fracture healing models, where topology optimization and polar moment of area constraint have not been introduced earlier.
Collapse
|
42
|
Strollo F, Gentile S, Strollo G, Mambro A, Vernikos J. Recent Progress in Space Physiology and Aging. Front Physiol 2018; 9:1551. [PMID: 30483144 PMCID: PMC6240610 DOI: 10.3389/fphys.2018.01551] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/16/2018] [Indexed: 12/17/2022] Open
Abstract
Astronauts coming back from long-term space missions present with different health problems potentially affecting mission performance, involving all functional systems and organs and closely resembling those found in the elderly. This review points out the most recent advances in the literature in areas of expertise in which specific research groups were particularly creative, and as they relate to aging and to possible benefits on Earth for disabled people. The update of new findings and approaches in space research refers especially to neuro-immuno-endocrine-metabolic interactions, optic nerve edema, motion sickness and muscle-tendon-bone interplay and aims at providing the curious - and even possibly naïve young researchers – with a source of inspiration and of creative ideas for translational research.
Collapse
Affiliation(s)
| | - Sandro Gentile
- Campania University "Luigi Vanvitelli" and Nefrocenter Research Network, Naples, Italy
| | | | - Andrea Mambro
- Anesthesiology and Resuscitation Unit, "Misercordia" Hospital, Grosseto, Italy
| | | |
Collapse
|
43
|
|