1
|
Chermside-Scabbo CJ, Shuster JT, Erdmann-Gilmore P, Tycksen E, Zhang Q, Townsend RR, Silva MJ. A proteomics approach to study mouse long bones: examining baseline differences and mechanical loading-induced bone formation in young-adult and old mice. Aging (Albany NY) 2024; 16:12726-12768. [PMID: 39400554 PMCID: PMC11501390 DOI: 10.18632/aging.206131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
With aging, bone mass declines and the anabolic effects of skeletal loading diminish. While much research has focused on gene transcription, how bone ages and loses its mechanoresponsiveness at the protein level remains unclear. We developed a novel proteomics approach and performed a paired mass spectrometry and RNA-seq analysis on tibias from young-adult (5-month) and old (22-month) mice. We report the first correlation estimate between the bone proteome and transcriptome (Spearman ρ = 0.40), which is in line with other tissues but indicates that a relatively low amount of variation in protein levels is explained by the variation in transcript levels. Of 71 shared targets that differed with age, eight were associated with bone mineral density in previous GWAS, including understudied targets Asrgl1 and Timp2. We used complementary RNA in situ hybridization to confirm that Asrgl1 and Timp2 had reduced expression in osteoblasts/osteocytes in old bones. We also found evidence for reduced TGF-beta signaling with aging, in particular Tgfb2. Next, we defined proteomic changes following mechanical loading. At the protein level, bone differed more with age than with loading, and aged bone had fewer loading-induced changes. Overall, our findings underscore the need for complementary protein-level assays in skeletal biology research.
Collapse
Affiliation(s)
- Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John T. Shuster
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Petra Erdmann-Gilmore
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric Tycksen
- Department of Genetics, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Qiang Zhang
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - R. Reid Townsend
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
2
|
Horasan M, Verner KA, Yang H, Main RP, Nauman EA. Computationally derived endosteal strain and strain gradients correlate with increased bone formation in an axially loaded murine tibia model. J Mech Behav Biomed Mater 2024; 160:106761. [PMID: 39388844 DOI: 10.1016/j.jmbbm.2024.106761] [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: 08/05/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/12/2024]
Abstract
Osteoporosis is a common metabolic bone disorder characterized by low bone mass and microstructural degradation of bone tissue due to a derailed bone remodeling process. A deeper understanding of the mechanobiological phenomena that modulate the bone remodeling response to mechanical loading in a healthy bone is crucial to develop treatments. Rodent models have provided invaluable insight into the mechanobiological mechanisms regulating bone adaptation in response to dynamic mechanic stimuli. This study sheds light on these aspects by means of assessing the mechanical environment of the cortical and cancellous tissue to in vivo dynamic compressive loading within the mouse tibia using microCT-based finite element model in combination with diaphyseal strain gauge measures. Additionally, this work describes the relation between the mid-diaphyseal strains and strain gradients from the finite element analysis and bone formation measures from time-lapse in vivo tibial loading with a fluorochrome-derived histomorphometry analysis. The mouse tibial loading model demonstrated that cancellous strains were lower than those in the midshaft cortical bone. Sensitivity analyses demonstrated that the material property of cortical bone was the most significant model parameter. The computationally-modeled strains and strain gradients correlated significantly to the histologically-measured bone formation thickness at the mid-diaphyseal cross-section of the mouse tibia.
Collapse
Affiliation(s)
- Murat Horasan
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA; Department of Mechanical Engineering, Aksaray University, Aksaray, Turkey.
| | - Kari A Verner
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Explico Engineering, Denver, CO, USA.
| | - Haisheng Yang
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA; Department of Biomedical Engineering, Beijing University of Technology, Beijing, China.
| | - Russell P Main
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA
| | - Eric A Nauman
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
3
|
Barak MM. Cortical and Trabecular Bone Modeling and Implications for Bone Functional Adaptation in the Mammalian Tibia. Bioengineering (Basel) 2024; 11:514. [PMID: 38790379 PMCID: PMC11118124 DOI: 10.3390/bioengineering11050514] [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: 04/23/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Bone modeling involves the addition of bone material through osteoblast-mediated deposition or the removal of bone material via osteoclast-mediated resorption in response to perceived changes in loads by osteocytes. This process is characterized by the independent occurrence of deposition and resorption, which can take place simultaneously at different locations within the bone due to variations in stress levels across its different regions. The principle of bone functional adaptation states that cortical and trabecular bone tissues will respond to mechanical stimuli by adjusting (i.e., bone modeling) their morphology and architecture to mechanically improve their mechanical function in line with the habitual in vivo loading direction. This principle is relevant to various research areas, such as the development of improved orthopedic implants, preventative medicine for osteopenic elderly patients, and the investigation of locomotion behavior in extinct species. In the present review, the mammalian tibia is used as an example to explore cortical and trabecular bone modeling and to examine its implications for the functional adaptation of bones. Following a short introduction and an exposition on characteristics of mechanical stimuli that influence bone modeling, a detailed critical appraisal of the literature on cortical and trabecular bone modeling and bone functional adaptation is given. By synthesizing key findings from studies involving small mammals (rodents), large mammals, and humans, it is shown that examining both cortical and trabecular bone structures is essential for understanding bone functional adaptation. A combined approach can provide a more comprehensive understanding of this significant physiological phenomenon, as each structure contributes uniquely to the phenomenon.
Collapse
Affiliation(s)
- Meir M Barak
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY 11548, USA
| |
Collapse
|
4
|
A L, Elsen R, Nayak S. Artificial Intelligence-Based 3D Printing Strategies for Bone Scaffold Fabrication and Its Application in Preclinical and Clinical Investigations. ACS Biomater Sci Eng 2024; 10:677-696. [PMID: 38252807 DOI: 10.1021/acsbiomaterials.3c01368] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
3D printing has become increasingly popular in the field of bone tissue engineering. However, the mechanical properties, biocompatibility, and porosity of the 3D printed bone scaffolds are major requirements for tissue regeneration and implantation as well. Designing the scaffold architecture in accordance with the need to create better mechanical and biological stimuli is necessary to achieve unique scaffold properties. To accomplish this, different 3D designing strategies can be utilized with the help of the scaffold design library and artificial intelligence (AI). The implementation of AI to assist the 3D printing process can enable it to predict, adapt, and control the parameters on its own, which lowers the risk of errors. This Review emphasizes 3D design and fabrication of bone scaffold using different materials and the use of AI-aided 3D printing strategies. Also, the adaption of AI to 3D printing helps to develop patient-specific scaffolds based on different requirements, thus providing feedback and adequate data for reproducibility, which can be improvised in the future. These printed scaffolds can also serve as an alternative to preclinical animal test models to cut costs and prevent immunological interference.
Collapse
Affiliation(s)
- Logeshwaran A
- School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Katpadi, Vellore, Tamil Nadu 632014, India
| | - Renold Elsen
- School of Mechanical Engineering, Vellore Institute of Technology (VIT), Katpadi, Vellore, Tamil Nadu 632014, India
| | - Sunita Nayak
- School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Katpadi, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
5
|
Farage-O’Reilly SM, Cheong VS, Pickering E, Pivonka P, Bellantuono I, Kadirkamanathan V, Dall’Ara E. The loading direction dramatically affects the mechanical properties of the mouse tibia. Front Bioeng Biotechnol 2024; 12:1335955. [PMID: 38380263 PMCID: PMC10877372 DOI: 10.3389/fbioe.2024.1335955] [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: 11/09/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
Introduction: The in vivo tibial loading mouse model has been extensively used to evaluate bone adaptation in the tibia after mechanical loading treatment. However, there is a prevailing assumption that the load is applied axially to the tibia. The aim of this in silico study was to evaluate how much the apparent mechanical properties of the mouse tibia are affected by the loading direction, by using a validated micro-finite element (micro-FE) model of mice which have been ovariectomized and exposed to external mechanical loading over a two-week period. Methods: Longitudinal micro-computed tomography (micro-CT) images were taken of the tibiae of eleven ovariectomized mice at ages 18 and 20 weeks. Six of the mice underwent a mechanical loading treatment at age 19 weeks. Micro-FE models were generated, based on the segmented micro-CT images. Three models using unitary loads were linearly combined to simulate a range of loading directions, generated as a function of the angle from the inferior-superior axis (θ, 0°-30° range, 5° steps) and the angle from the anterior-posterior axis (ϕ, 0°: anterior axis, positive anticlockwise, 0°-355° range, 5° steps). The minimum principal strain was calculated and used to estimate the failure load, by linearly scaling the strain until 10% of the nodes reached the critical strain level of -14,420 με. The apparent bone stiffness was calculated as the ratio between the axial applied force and the average displacement along the longitudinal direction, for the loaded nodes. Results: The results demonstrated a high sensitivity of the mouse tibia to the loading direction across all groups and time points. Higher failure loads were found for several loading directions (θ = 10°, ϕ 205°-210°) than for the nominal axial case (θ = 0°, ϕ = 0°), highlighting adaptation of the bone for loading directions far from the nominal axial one. Conclusion: These results suggest that in studies which use mouse tibia, the loading direction can significantly impact the failure load. Thus, the magnitude and direction of the applied load should be well controlled during the experiments.
Collapse
Affiliation(s)
- Saira Mary Farage-O’Reilly
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
- Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Vee San Cheong
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Future Health Technologies Programme, Singapore-ETH Centre, Singapore, Singapore
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Edmund Pickering
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| | - Peter Pivonka
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ilaria Bellantuono
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
- Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Visakan Kadirkamanathan
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Enrico Dall’Ara
- Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
- Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
6
|
Zhang L, Guan Q, Wang Z, Feng J, Zou J, Gao B. Consequences of Aging on Bone. Aging Dis 2023:AD.2023.1115. [PMID: 38029404 DOI: 10.14336/ad.2023.1115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023] Open
Abstract
With the aging of the global population, the incidence of musculoskeletal diseases has been increasing, seriously affecting people's health. As people age, the microenvironment within skeleton favors bone resorption and inhibits bone formation, accompanied by bone marrow fat accumulation and multiple cellular senescence. Specifically, skeletal stem/stromal cells (SSCs) during aging tend to undergo adipogenesis rather than osteogenesis. Meanwhile, osteoblasts, as well as osteocytes, showed increased apoptosis, decreased quantity, and multiple functional limitations including impaired mechanical sensing, intercellular modulation, and exosome secretion. Also, the bone resorption function of macrophage-lineage cells (including osteoclasts and preosteoclasts) was significantly enhanced, as well as impaired vascularization and innervation. In this study, we systematically reviewed the effect of aging on bone and the within microenvironment (including skeletal cells as well as their intracellular structure variations, vascular structures, innervation, marrow fat distribution, and lymphatic system) caused by aging, and mechanisms of osteoimmune regulation of the bone environment in the aging state, and the causal relationship with multiple musculoskeletal diseases in addition with their potential therapeutic strategy.
Collapse
Affiliation(s)
- Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Qiao Guan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Zhikun Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jie Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
7
|
Sarazin BA, Liu B, Goldman E, Whitefield AN, Lynch ME. Bone-homing metastatic breast cancer cells impair osteocytes' mechanoresponse in a 3D loading model. Heliyon 2023; 9:e20248. [PMID: 37767467 PMCID: PMC10520780 DOI: 10.1016/j.heliyon.2023.e20248] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/23/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer predominantly metastasizes to the skeleton. Mechanical loading is reliably anabolic in bone, and also inhibits bone metastatic tumor formation and bone loss in vivo. To study the underlying mechanisms, we developed a 3D culture model for osteocytes, the primary bone mechanosensor. We verified that MLO-Y4s responded to perfusion by reducing their rankl and rankl:opg gene expression. We next cultured MLO-Y4s with tumor-conditioned media (TCM) collected from human breast cancer cells (MDA-MB-231s) and a corresponding bone-homing subclone to test the impacts on osteocytes' mechanosensation. We found that TCM from the bone-homing subclone was more detrimental to MLO-Y4 growth and viability, and it abrogated loading-induced changes to rankl:opg. Our studies demonstrate that MLO-Y4s, including their mechanoresponse to perfusion, were more negatively impacted by soluble factors from bone-homing breast cancer cells compared to those from parental cells.
Collapse
Affiliation(s)
- Blayne A. Sarazin
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Boyuan Liu
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Elaine Goldman
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Ashlyn N. Whitefield
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Maureen E. Lynch
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80309, USA
| |
Collapse
|
8
|
Li X, Zhang C, Bowman HH, Stambough JB, Stronach BM, Mears SC, Barnes LC, Ambrogini E, Xiong J. Piezo1 opposes age-associated cortical bone loss. Aging Cell 2023:e13846. [PMID: 37147884 DOI: 10.1111/acel.13846] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 05/07/2023] Open
Abstract
As we age, our bones undergo a process of loss, often accompanied by muscle weakness and reduced physical activity. This is exacerbated by decreased responsiveness to mechanical stimulation in aged skeleton, leading to the hypothesis that decreased mechanical stimulation plays an important role in age-related bone loss. Piezo1, a mechanosensitive ion channel, is critical for bone homeostasis and mechanotransduction. Here, we observed a decrease in Piezo1 expression with age in both murine and human cortical bone. Furthermore, loss of Piezo1 in osteoblasts and osteocytes resulted in an increase in age-associated cortical bone loss compared to control mice. The loss of cortical bone was due to an expansion of the endosteal perimeter resulting from increased endocortical resorption. In addition, expression of Tnfrsf11b, encoding anti-osteoclastogenic protein OPG, decreases with Piezo1 in vitro and in vivo in bone cells, suggesting that Piezo1 suppresses osteoclast formation by promoting Tnfrsf11b expression. Our results highlight the importance of Piezo1-mediated mechanical signaling in protecting against age-associated cortical bone loss by inhibiting bone resorption in mice.
Collapse
Affiliation(s)
- Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Connie Zhang
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Hayden H Bowman
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jeffrey B Stambough
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Benjamin M Stronach
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Simon C Mears
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Lowry C Barnes
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Elena Ambrogini
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
9
|
Xu L, Hu YJ, Peng Y, Wang Z, Wang J, Lu WW, Tang B, Guo XE. Early zoledronate treatment inhibits subchondral bone microstructural changes in skeletally-mature, ACL-transected canine knees. Bone 2023; 167:116638. [PMID: 36464243 DOI: 10.1016/j.bone.2022.116638] [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: 09/04/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Anterior cruciate ligament (ACL) tear leads to post-traumatic osteoarthritis (PTOA), a significant clinical burden worldwide that currently has no cure. Recent studies suggest a role of subchondral bone adaptations in the development of PTOA. Particularly, microstructural changes in the rod-and-plate microstructure of subchondral bone may precede and contribute to OA progression. In this study, we quantified microstructural changes in subchondral trabecular rods and plates after ACL-transection for the first time in the well-established preclinical canine model of PTOA and investigated the therapeutic potentials of a bisphosphonate (zoledronate) and NSAID treatment (meloxicam). Unilateral hindlimb ACL transection was performed on skeletally-mature (2-year-old, N = 20) and juvenile (10-month-old, N = 20) male beagles. Animals were assigned to 4 groups (N = 5): ACLT, un-operated control, ACLT with zoledronate, and ACLT with meloxicam treatment. Subchondral bone microstructure was evaluated by micro-computed tomography and cartilage integrity was evaluated histologically. We found that ACL-induced subchondral bone changes depended on skeletal maturity of animals. In mature animals, significant loss of trabecular plates that resulted in reduced PR ratio occurred at Month 1 and persisted until Month 8. Zoledronate treatment prevented trabecular plate loss while meloxicam treatment did not. Whether cartilage degeneration is also attenuated warrants further investigation. In juvenile animals that have not reached skeletal maturity, transient changes in trabecular plate and rod microstructure occurred at Month 3 but not Month 9. Neither zoledronate nor meloxicam treatment attenuated bone microstructural changes or cartilage damages. Findings from this study suggest that early inhibition of bone resorption by bisphosphonate after injury may be a promising therapeutic approach to prevent alterations in subchondral bone microstructure associated with PTOA. Our results further demonstrate that pathogenesis of PTOA may differ between adolescent and adult patients and therefore require distinct management strategies.
Collapse
Affiliation(s)
- Lei Xu
- Department of Biomedical Engineering, the Southern University of Science and Technology, Shenzhen, PR China; Department of Orthopeadics and Traumatology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong; Department of Orthopeadics and Traumatology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, PR China
| | - Yizhong Jenny Hu
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Ying Peng
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Zexi Wang
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jingyi Wang
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - W William Lu
- Department of Orthopeadics and Traumatology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Bin Tang
- Department of Biomedical Engineering, the Southern University of Science and Technology, Shenzhen, PR China
| | - X Edward Guo
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| |
Collapse
|
10
|
Xu X, Yang H, Bullock WA, Gallant MA, Ohlsson C, Bellido TM, Main RP. Osteocyte Estrogen Receptor β (Ot-ERβ) Regulates Bone Turnover and Skeletal Adaptive Response to Mechanical Loading Differently in Male and Female Growing and Adult Mice. J Bone Miner Res 2023; 38:186-197. [PMID: 36321245 PMCID: PMC10108310 DOI: 10.1002/jbmr.4731] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Age-related bone loss is a failure of balanced bone turnover and diminished skeletal mechanoadaptation. Estrogen receptors, ERα and ERβ, play critical roles in osteoprotective regulation activated by estrogen and mechanical signals. Previous studies mainly focused on ERα and showed that osteocyte-ERα (Ot-ERα) regulated trabecular, but not cortical bone, and played a minor role in load-induced cortical adaptation. However, the role of Ot-ERβ in bone mass regulation remains unrevealed. To address this issue, we characterized bone (re)modeling and gene expression in male and female mice with Ot-ERβ deletion (ERβ-dOT) and littermate control (LC) at 10 weeks (young) or 28 weeks (adult) of age, as well as their responses to in vivo tibial compressive loading. Increased cancellous bone mass appeared in the L4 vertebral body of young male ERβ-dOT mice. At the same time, femoral cortical bone gene expression showed signs consistent with elevated osteoblast and osteoclast activities (type-I collagen, Cat K, RANKL). Upregulated androgen receptor (AR) expression was observed in young male ERβ-dOT mice relative to LC, suggesting a compensatory effect of testosterone on male bone protection. In contrast, bone mass in L4 decreased in adult male ERβ-dOT mice, attributed to potentially increased bone resorption activity (Cat K) with no change in bone formation. There was no effect of ERβ-dOT on bone mass or gene expression in female mice. Sex-dependent regulation of Ot-ERβ also appeared in load-induced cortical responsiveness. Young female ERβ-dOT mice showed an enhanced tibial cortical anabolic adaptation compared with LC. In contrast, an attenuated cortical anabolic response presented at the proximal tibia in male ERβ-dOT mice at both ages. For the first time, our findings suggest that Ot-ERβ regulates bone (re)modeling and the response to mechanical signals through different mechanisms in males and females. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Xiaoyu Xu
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteINUSA
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| | - Haisheng Yang
- Department of Biomedical Engineering, Faculty of Environment and LifeBeijing University of TechnologyBeijingChina
| | | | - Maxim A. Gallant
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical NutritionInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Drug TreatmentSahlgrenska University HospitalGothenburgSweden
| | - Teresita M. Bellido
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockARUSA
| | - Russell P. Main
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteINUSA
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
11
|
Meslier QA, DiMauro N, Somanchi P, Nano S, Shefelbine SJ. Manipulating load-induced fluid flow in vivo to promote bone adaptation. Bone 2022; 165:116547. [PMID: 36113842 DOI: 10.1016/j.bone.2022.116547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/02/2022]
Abstract
Mechanical stimulation is critical to maintaining bone mass and strength. Strain has been commonly thought of as the mechanical stimulus driving bone adaptation. However, numerous studies have hypothesized that fluid flow in the lacunar-canalicular system plays a role in mechanoadaptation. The role of fluid flow compared to strain magnitude on bone remodeling has yet to be characterized. This study aimed to determine the contribution of fluid flow velocity compared to strain on bone adaptation. We used finite element modeling to design in vivo experiments, manipulating strain and fluid flow contributions. Using a uniaxial compression tibia model in mice, we demonstrated that high fluid flow velocity results in significant bone adaptation even under low strain magnitude. In contrast, high strain magnitude paired with low fluid velocity does not trigger a bone response. These findings support previous hypotheses stating that fluid flow is the principal mechanical stimulus driving bone adaptation. Moreover, they give new insights regarding bone adaptative response and provide new pathways toward treatment against age-related mechanosensitivity loss in bone.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Nicole DiMauro
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Priya Somanchi
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Sarah Nano
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
12
|
Ayobami OO, Goldring SR, Goldring MB, Wright TM, van der Meulen MCH. Contribution of joint tissue properties to load-induced osteoarthritis. Bone Rep 2022; 17:101602. [PMID: 35899096 PMCID: PMC9309407 DOI: 10.1016/j.bonr.2022.101602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 06/29/2022] [Accepted: 07/11/2022] [Indexed: 11/27/2022] Open
Abstract
Objective Clinical evidence suggests that abnormal mechanical forces play a major role in the initiation and progression of osteoarthritis (OA). However, few studies have examined the mechanical environment that leads to disease. Thus, using a mouse tibial loading model, we quantified the cartilage contact stresses and examined the effects of altering tissue material properties on joint stresses during loading. Design Using a discrete element model (DEA) in conjunction with joint kinematics data from a murine knee joint compression model, the magnitude and distribution of contact stresses in the tibial cartilage during joint loading were quantified at levels ranging from 0 to 9 N in 1 N increments. In addition, a simplified finite element (FEA) contact model was developed to simulate the knee joint, and parametric analyses were conducted to investigate the effects of altering bone and cartilage material properties on joint stresses during compressive loading. Results As loading increased, the peak contact pressures were sufficient to induce fibrillations on the cartilage surfaces. The computed areas of peak contact pressures correlated with experimentally defined areas of highest cartilage damage. Only alterations in cartilage properties and geometry caused large changes in cartilage contact pressures. However, changes in both bone and cartilage material properties resulted in significant changes in stresses induced in the bone during compressive loading. Conclusions The level of mechanical stress induced by compressive tibial loading directly correlated with areas of biological change observed in the mouse knee joint. These results, taken together with the parametric analyses, are the first to demonstrate both experimentally and computationally that the tibial loading model is a useful preclinical platform with which to predict and study the effects of modulating bone and/or cartilage properties on attenuating OA progression. Given the direct correlation between computational modeling and experimental results, the effects of tissue-modifying treatments may be predicted prior to in vivo experimentation, allowing for novel therapeutics to be developed.
Collapse
Affiliation(s)
- Olufunmilayo O Ayobami
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Steven R Goldring
- Research Division, Hospital for Special Surgery, New York, NY, United States of America
| | - Mary B Goldring
- Research Division, Hospital for Special Surgery, New York, NY, United States of America
| | - Timothy M Wright
- Research Division, Hospital for Special Surgery, New York, NY, United States of America
| | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America.,Research Division, Hospital for Special Surgery, New York, NY, United States of America
| |
Collapse
|
13
|
Zeng Y, Riquelme MA, Hua R, Zhang J, Acosta FM, Gu S, Jiang JX. Mechanosensitive piezo1 calcium channel activates connexin 43 hemichannels through PI3K signaling pathway in bone. Cell Biosci 2022; 12:191. [PMID: 36457052 PMCID: PMC9716748 DOI: 10.1186/s13578-022-00929-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Mechanical loading promotes bone formation and osteocytes are a major mechanosensory cell in the bone. Both Piezo1 channels and connexin 43 hemichannels (Cx43 HCs) in osteocytes are important players in mechanotransduction and anabolic function by mechanical loading. However, the mechanism underlying mechanotransduction involving Piezo1 channels and Cx43 HCs in osteocytes and bone remains unknown. RESULTS We showed that, like mechanical loading, Piezo1 specific agonist Yoda1 was able to increase intracellular Ca2+ signaling and activate Cx43 HCs, while Yoda1 antagonist Dooku1 inhibited Ca2+ and Cx43 HC activation induced by both mechanical loading and Yoda1. Moreover, the intracellular Ca2+ signal activated by Yoda1 was reduced by the inhibition of Cx43 HCs and pannexin1 (Panx1) channels, as well as ATP-P2X receptor signaling. Piezo1 and Cx43 HCs were co-localized on the osteocyte cell surface, and Yoda1-activated PI3K-Akt signaling regulated the opening of Cx43 HCs. Furthermore, Cx43 HCs opening by mechanical loading on tibias was ablated by inhibition of Piezo1 activation in vivo. CONCLUSION We demonstrated that upon mechanical stress, increased intracellular Ca2+ activated by Piezo1 regulates the opening of HCs through PI3K-Akt and opened Cx43 HCs, along with Panx1 channels, and ATP-P2X signaling sustain the intracellular Ca2+ signal, leading to bone anabolic function.
Collapse
Affiliation(s)
- Yan Zeng
- grid.267309.90000 0001 0629 5880Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA ,grid.452708.c0000 0004 1803 0208The Second Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Manuel A. Riquelme
- grid.267309.90000 0001 0629 5880Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
| | - Rui Hua
- grid.267309.90000 0001 0629 5880Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
| | - Jingruo Zhang
- grid.267309.90000 0001 0629 5880Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
| | - Francisca M. Acosta
- grid.267309.90000 0001 0629 5880Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
| | - Sumin Gu
- grid.267309.90000 0001 0629 5880Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
| | - Jean X. Jiang
- grid.267309.90000 0001 0629 5880Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
| |
Collapse
|
14
|
Chlebek C, Moore JA, Ross FP, van der Meulen MCH. Molecular Identification of Spatially Distinct Anabolic Responses to Mechanical Loading in Murine Cortical Bone. J Bone Miner Res 2022; 37:2277-2287. [PMID: 36054133 DOI: 10.1002/jbmr.4686] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/05/2022] [Accepted: 08/20/2022] [Indexed: 11/08/2022]
Abstract
Osteoporosis affects over 200 million women worldwide, one-third of whom are predicted to suffer from an osteoporotic fracture in their lifetime. The most promising anabolic drugs involve administration of expensive antibodies. Because mechanical loading stimulates bone formation, our current data, using a mouse model, replicates the anabolic effects of loading in humans and may identify novel pathways amenable to oral treatment. Murine tibial compression produces axially varying deformations along the cortical bone, inducing highest strains at the mid-diaphysis and lowest at the metaphyseal shell. To test the hypothesis that load-induced transcriptomic responses at different axial locations of cortical bone would vary as a function of strain magnitude, we loaded the left tibias of 10-week-old female C57Bl/6 mice in vivo in compression, with contralateral limbs as controls. Animals were euthanized at 1, 3, or 24 hours post-loading or loaded for 1 week (n = 4-5/group). Bone marrow and cancellous bone were removed, cortical bone was segmented into the metaphyseal shell, proximal diaphysis, and mid-diaphysis, and load-induced differential gene expression and enriched biological processes were examined for the three segments. At each time point, the mid-diaphysis (highest strain) had the greatest transcriptomic response. Similarly, biological processes regulating bone formation and turnover increased earlier and to the greatest extent at the mid-diaphysis. Higher strain induced greater levels of osteoblast and osteocyte genes, whereas expression was lower in osteoclasts. Among the top differentially expressed genes at 24-hours post-loading, 17 had known functions in bone biology, of which 12 were present only in osteoblasts, 3 exclusively in osteoclasts, and 2 were present in both cell types. Based on these results, we conclude that murine tibial loading induces spatially unique transcriptomic responses correlating with strain magnitude in cortical bone. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Carolyn Chlebek
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jacob A Moore
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | | | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
15
|
Effects of Dietary Protein Source and Quantity on Bone Morphology and Body Composition Following a High-Protein Weight-Loss Diet in a Rat Model for Postmenopausal Obesity. Nutrients 2022; 14:nu14112262. [PMID: 35684064 PMCID: PMC9183012 DOI: 10.3390/nu14112262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022] Open
Abstract
Higher protein (>30% of total energy, HP)-energy restriction (HP-ER) diets are an effective means to improve body composition and metabolic health. However, weight loss (WL) is associated with bone loss, and the impact of HP-ER diets on bone is mixed and controversial. Recent evidence suggests conflicting outcomes may stem from differences in age, hormonal status, and the predominant source of dietary protein consumed. Therefore, this study investigated the effect of four 12-week energy restriction (ER) diets varying in predominate protein source (beef, milk, soy, casein) and protein quantity (normal protein, NP 15% vs. high, 35%) on bone and body composition outcomes in 32-week-old obese, ovariectomized female rats. Overall, ER decreased body weight, bone quantity (aBMD, aBMC), bone microarchitecture, and body composition parameters. WL was greater with the NP vs. HP-beef and HP-soy diets, and muscle area decreased only with the NP diet. The HP-beef diet exacerbated WL-induced bone loss (increased trabecular separation and endocortical bone formation rates, lower bone retention and trabecular BMC, and more rod-like trabeculae) compared to the HP-soy diet. The HP-milk diet did not augment WL-induced bone loss. Results suggest that specific protein source recommendations may be needed to attenuate the adverse alterations in bone quality following an HP-ER diet in a model of postmenopausal obesity.
Collapse
|
16
|
Wang H, Du T, Li R, Main RP, Yang H. Interactive effects of various loading parameters on the fluid dynamics within the lacunar-canalicular system for a single osteocyte. Bone 2022; 158:116367. [PMID: 35181573 DOI: 10.1016/j.bone.2022.116367] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 12/26/2022]
Abstract
The osteocyte lacunar-canalicular system (LCS) serves as a mechanotransductive core where external loading applied to the skeleton is transduced into mechanical signals (e.g., fluid shear) that can be sensed by mechanosensors (osteocytes). The fluid velocity and shear stress within the LCS are affected by various loading parameters. However, the interactive effect of distinct loading parameters on the velocity and shear stress in the LCS remains unclear. To address this issue, we developed a multiscale modeling approach, combining a poroelastic finite element (FE) model with a single osteocytic LCS unit model to calculate the flow velocity and shear stress within the LCS. Next, a sensitivity analysis was performed to investigate individual and interactive effects of strain magnitude, strain rate, number of cycles, and intervening short rests between loading cycles on the velocity and shear stress around the osteocyte. Lastly, we developed a relatively simple regression model to predict those outcomes. Our results demonstrated that the strain magnitude or rate alone were the main factors affecting the velocity and shear stress; however, the combination of these two was not directly additive, and addition of a short rest between cycles could enhance the combination of these two related factors. These results show highly interactive effects of distinct loading parameters on fluid velocity and shear stress in the LCS. Specifically, our results suggest that an enhanced fluid dynamics environment in the LCS can be achieved with a brief number of load cycles combined with short rest insertion and high strain magnitude and rate.
Collapse
Affiliation(s)
- Huiru Wang
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Tianming Du
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Rui Li
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Russell P Main
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, IN, USA; Weldon School of Biomedical Engineering, Purdue University, IN, USA
| | - Haisheng Yang
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
17
|
Rooney AM, Ayobami OO, Kelly NH, Schimenti JC, Ross FP, van der Meulen MCH. Bone mass and adaptation to mechanical loading are sexually dimorphic in adult osteoblast-specific ERα knockout mice. Bone 2022; 158:116349. [PMID: 35123146 DOI: 10.1016/j.bone.2022.116349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/20/2022] [Accepted: 01/31/2022] [Indexed: 12/14/2022]
Abstract
Estrogen receptor-alpha (ERα) regulates bone mass and is implicated in bone tissue's response to mechanical loading. The effects of ERα deletion in mice depend on sex, anatomical location, and the cellular stage at which ERα is removed. Few studies have investigated the effect of age on the role of ERα in skeletal maintenance and functional adaptation. We previously demonstrated that bone mass and adaptation to loading were altered in growing 10-week-old female and male mice lacking ERα in mature osteoblasts and osteocytes (pOC-ERαKO). Here our goal was to determine the effects of ERα and mechanical loading in skeletally-mature adult mice. We subjected 26-week-old skeletally-mature adult pOC-ERαKO and littermate control (LC) mice of both sexes to two weeks of in vivo cyclic tibial loading. ERα deletion in male mice did not alter bone mass or the response to loading. Adult female pOC-ERαKO mice had reduced cancellous and cortical bone mass and increased adaptation to high-magnitude mechanical loading compared to LC mice. Thus, ERα deletion from mature osteoblasts reduced the bone mass and increased the mechanoadaptation of adult female but not male mice. Additionally, compared to our previous work in young mice, adult female mice had greatly reduced mechanoadaptation and adult male mice retained most of their mechanoadaptation with age.
Collapse
Affiliation(s)
- Amanda M Rooney
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Olufunmilayo O Ayobami
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Natalie H Kelly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - John C Schimenti
- College of Veterinary Medicine, Cornell University, Ithaca 14853, NY, USA.
| | - F Patrick Ross
- Research Division, Hospital for Special Surgery, New York, NY 10021, USA.
| | - Marjolein C H van der Meulen
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Research Division, Hospital for Special Surgery, New York, NY 10021, USA.
| |
Collapse
|
18
|
Cui J, Shibata Y, Zhu T, Zhou J, Zhang J. Osteocytes in bone aging: Advances, challenges, and future perspectives. Ageing Res Rev 2022; 77:101608. [PMID: 35283289 DOI: 10.1016/j.arr.2022.101608] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Osteocytes play a critical role in maintaining bone homeostasis and in regulating skeletal response to hormones and mechanical loading. Substantial evidence have demonstrated that osteocytes and their lacunae exhibit morphological changes in aged bone, indicating the underlying involvement of osteocytes in bone aging. Notably, recent studies have deciphered aged osteocytes to have characteristics such as impaired mechanosensitivity, accumulated cellular senescence, dysfunctional perilacunar/canalicular remodeling, and degenerated lacuna-canalicular network. However, detailed molecular mechanisms of osteocytes remain unclear. Nonetheless, osteocyte transcriptomes analyzed via advanced RNA sequencing (RNA-seq) techniques have identified several bone aging-related genes and signaling pathways, such as Wnt, Bmp/TGF, and Jak-STAT. Moreover, inflammation, immune dysfunction, energy shortage, and impaired hormone responses possibly affect osteocytes in age-related bone deterioration. In this review, we summarize the hallmarks of aging bone and osteocytes and discuss osteocytic mechanisms in age-related bone loss and impaired bone quality. Furthermore, we provide insights into the challenges faced and their possible solutions when investigating osteocyte transcriptomes. We also highlight that single-cell RNA-seq can decode transcriptomic messages in aged osteocytes; therefore, this technique can promote novel single cell-based investigations in osteocytes once a well-established standardized protocol specific for osteocytes is developed. Interestingly, improved understanding of osteocytic mechanisms have helped identify promising targets and effective therapies for aging-related osteoporosis and fragile fractures.
Collapse
|
19
|
Goldsmith M, Crooks SD, Condon SF, Willie BM, Komarova SV. Bone strength and composition in spacefaring rodents: systematic review and meta-analysis. NPJ Microgravity 2022; 8:10. [PMID: 35418128 PMCID: PMC9008045 DOI: 10.1038/s41526-022-00195-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 03/04/2022] [Indexed: 11/09/2022] Open
Abstract
Studying the effects of space travel on bone of experimental animals provides unique advantages, including the ability to perform post-mortem analysis and mechanical testing. To synthesize the available data to assess how much and how consistently bone strength and composition parameters are affected by spaceflight, we systematically identified studies reporting bone health in spacefaring animals from Medline, Embase, Web of Science, BIOSIS, and NASA Technical reports. Previously, we reported the effect of spaceflight on bone architecture and turnover in rodents and primates. For this study, we selected 28 articles reporting bone strength and composition in 60 rats and 60 mice from 17 space missions ranging from 7 to 33 days in duration. Whole bone mechanical indices were significantly decreased in spaceflight rodents, with the percent difference between spaceflight and ground control animals for maximum load of −15.24% [Confidence interval: −22.32, −8.17]. Bone mineral density and calcium content were significantly decreased in spaceflight rodents by −3.13% [−4.96, −1.29] and −1.75% [−2.97, −0.52] respectively. Thus, large deficits in bone architecture (6% loss in cortical area identified in a previous study) as well as changes in bone mass and tissue composition likely lead to bone strength reduction in spaceflight animals.
Collapse
Affiliation(s)
- Matthew Goldsmith
- Research Centre, Shriners Hospital for Children - Canada, Montréal, QC, Canada.,Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada
| | - Sequoia D Crooks
- Research Centre, Shriners Hospital for Children - Canada, Montréal, QC, Canada
| | - Sean F Condon
- Research Centre, Shriners Hospital for Children - Canada, Montréal, QC, Canada
| | - Bettina M Willie
- Research Centre, Shriners Hospital for Children - Canada, Montréal, QC, Canada.,Department of Pediatric Surgery, McGill University, Montréal, QC, Canada
| | - Svetlana V Komarova
- Research Centre, Shriners Hospital for Children - Canada, Montréal, QC, Canada. .,Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC, Canada.
| |
Collapse
|
20
|
Multibody Computer Model of the Entire Equine Forelimb Simulates Forces Causing Catastrophic Fractures of the Carpus during a Traditional Race. Animals (Basel) 2022; 12:ani12060737. [PMID: 35327134 PMCID: PMC8944875 DOI: 10.3390/ani12060737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/01/2022] [Accepted: 03/11/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Palios are traditional horseraces held in the main square of few Italian cities. Due to peculiar features of such circuits, adapted to the square architecture and thus characterized by tight curves and unconventional footing surface, horses involved are at particular risk of accidents. Prevention of catastrophic musculoskeletal injuries is a significant issue and matter of debate during these events. In particular, the negotiation of the curves in the city circuits is a significative concern. An experiment was set up to build a model of entire forelimb at the point of failure in the context of a turn comparable to that in a Palio circuit. The model was informed by live data and the output compared to post-mortem findings obtained from a horse that sustained a catastrophic fracture of the carpus during this competition. The objective of this study is to determine the magnitude and distribution of internal forces generated across the carpus under which the catastrophic injury has occurred and describe related post-mortem findings. Abstract A catastrophic fracture of the radial carpal bone experienced by a racehorse during a Palio race was analyzed. Computational modelling of the carpal joint at the point of failure informed by live data was generated using a multibody code for dynamics simulation. The circuit design in a turn, the speed of the animal and the surface characteristics were considered in the model. A macroscopic examination of the cartilage, micro-CT and histology were performed on the radio-carpal joint of the limb that sustained the fracture. The model predicted the points of contact forces generated at the level of the radio-carpal joint where the fracture occurred. Articular surfaces of the distal radius, together with the proximal articular surface of small carpal bones, exhibited diffuse wear lines, erosions of the articular cartilage and subchondral bone exposure. Even though the data in this study originated from a single fracture and further work will be required to validate this approach, this study highlights the potential correlation between elevated impact forces generated at the level of contact surfaces of the carpal joint during a turn and cartilage breakdown in the absence of pre-existing pathology. Computer modelling resulted in a useful tool to inversely calculate internal forces generated during specific conditions that cannot be reproduced in-vivo because of ethical concerns.
Collapse
|
21
|
Lai X, Chung R, Li Y, Liu XS, Wang L. Lactation alters fluid flow and solute transport in maternal skeleton: A multiscale modeling study on the effects of microstructural changes and loading frequency. Bone 2021; 151:116033. [PMID: 34102350 PMCID: PMC8276854 DOI: 10.1016/j.bone.2021.116033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/23/2021] [Accepted: 06/01/2021] [Indexed: 01/02/2023]
Abstract
The female skeleton undergoes significant material and ultrastructural changes to meet high calcium demands during reproduction and lactation. Through the peri-lacunar/canalicular remodeling (PLR), osteocytes actively resorb surrounding matrix and enlarge their lacunae and canaliculi during lactation, which are quickly reversed after weaning. How these changes alter the physicochemical environment of osteocytes, the most abundant and primary mechanosensing cells in bone, are not well understood. In this study, we developed a multiscale poroelastic modeling technique to investigate lactation-induced changes in stress, fluid pressurization, fluid flow, and solute transport across multiple length scales (whole bone, porous midshaft cortex, lacunar-canalicular pore system (LCS), and pericellular matrix (PCM) around osteocytes) in murine tibiae subjected to axial compression at 3 N peak load (~320 με) at 0.5, 2, or 4 Hz. Based on previously reported skeletal anatomical measurements from lactating and nulliparous mice, our models demonstrated that loading frequency, LCS porosity, and PCM density were major determinants of fluid and solute flows responsible for osteocyte mechanosensing, cell-cell signaling, and metabolism. When loaded at 0.5 Hz, lactation-induced LCS expansion and potential PCM reduction promoted solute transport and osteocyte mechanosensing via primary cilia, but suppressed mechanosensing via fluid shear and/or drag force on the cell membrane. Interestingly, loading at 2 or 4 Hz was found to overcome the mechanosensing deficits observed at 0.5 Hz and these counter effects became more pronounced at 4 Hz and with sparser PCM in the lactating bone. Synergistically, higher loading frequency (2, 4 Hz) and sparser PCM enhanced flow-mediated mechanosensing and diffusion/convection of nutrients and signaling molecules for osteocytes. In summary, lactation-induced structural changes alter the local environment of osteocytes in ways that favor metabolism, mechanosensing, and post-weaning recovery of maternal bone. Thus, osteocytes play a role in balancing the metabolic and mechanical functions of female skeleton during reproduction and lactation.
Collapse
Affiliation(s)
- Xiaohan Lai
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Rebecca Chung
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yihan Li
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiaowei Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, United States.
| |
Collapse
|
22
|
Li Y, de Bakker CMJ, Lai X, Zhao H, Parajuli A, Tseng WJ, Pei S, Meng T, Chung R, Wang L, Liu XS. Maternal bone adaptation to mechanical loading during pregnancy, lactation, and post-weaning recovery. Bone 2021; 151:116031. [PMID: 34098162 PMCID: PMC8504362 DOI: 10.1016/j.bone.2021.116031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/25/2021] [Accepted: 06/01/2021] [Indexed: 12/16/2022]
Abstract
The maternal skeleton undergoes dramatic bone loss during pregnancy and lactation, and substantial bone recovery post-weaning. The structural adaptations of maternal bone during reproduction and lactation exert a better protection of the mechanical integrity at the critical load-bearing sites, suggesting the importance of physiological load-bearing in regulating reproduction-induced skeletal alterations. Although it is suggested that physical exercise during pregnancy and breastfeeding improves women's physical and psychological well-being, its effects on maternal bone health remain unclear. Therefore, the objective of this study was to investigate the maternal bone adaptations to external mechanical loading during pregnancy, lactation, and post-weaning recovery. By utilizing an in vivo dynamic tibial loading protocol in a rat model, we demonstrated improved maternal cortical bone structure in response to dynamic loading at tibial midshaft, regardless of reproductive status. Notably, despite the minimal loading responses detected in the trabecular bone in virgins, rat bone during lactation experienced enhanced mechano-responsiveness in both trabecular and cortical bone compartments when compared to rats at other reproductive stages or age-matched virgins. Furthermore, our study showed that the lactation-induced elevation in osteocyte peri-lacunar/canalicular remodeling (PLR) activities led to enlarged osteocyte lacunae. This may result in alterations in interstitial fluid flow-mediated mechanical stimulation on osteocytes and an elevation in solute transport through the lacunar-canalicular system (LCS) during high-frequency dynamic loading, thus enhancing mechano-responsiveness of maternal bone during lactation. Taken together, findings from this study provide important insights into the relationship between reproduction- and lactation-induced skeletal changes and external mechanical loading, emphasizing the importance of weight-bearing exercise on maternal bone health during reproduction and postpartum.
Collapse
Affiliation(s)
- Yihan Li
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Chantal M J de Bakker
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Radiology, Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - Xiaohan Lai
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongbo Zhao
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ashutosh Parajuli
- Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Wei-Ju Tseng
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Shaopeng Pei
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Tan Meng
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rebecca Chung
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liyun Wang
- Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - X Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Lin CY, Kang JH. Mechanical Properties of Compact Bone Defined by the Stress-Strain Curve Measured Using Uniaxial Tensile Test: A Concise Review and Practical Guide. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4224. [PMID: 34361418 PMCID: PMC8347989 DOI: 10.3390/ma14154224] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/03/2022]
Abstract
Mechanical properties are crucial parameters for scaffold design for bone tissue engineering; therefore, it is important to understand the definitions of the mechanical properties of bones and relevant analysis methods, such that tissue engineers can use this information to properly design the mechanical properties of scaffolds for bone tissue engineering. The main purpose of this article is to provide a review and practical guide to understand and analyze the mechanical properties of compact bone that can be defined and extracted from the stress-strain curve measured using uniaxial tensile test until failure. The typical stress-strain curve of compact bone measured using uniaxial tensile test until failure is a bilinear, monotonically increasing curve. The associated mechanical properties can be obtained by analyzing this bilinear stress-strain curve. In this article, a computer programming code for analyzing the bilinear stress-strain curve of compact bone for quantifying the associated mechanical properties is provided, such that the readers can use this computer code to perform the analysis directly. In addition to being applied to compact bone, the information provided by this article can also be applied to quantify the mechanical properties of any material having a bilinear stress-strain curve, such as a whole bone, some metals and biomaterials. The information provided by this article can be applied by tissue engineers, such that they can have a reference to properly design the mechanical properties of scaffolds for bone tissue engineering. The information can also be applied by researchers in biomechanics and orthopedics to compare the mechanical properties of bones in different physiological or pathological conditions.
Collapse
Affiliation(s)
- Che-Yu Lin
- Institute of Applied Mechanics, College of Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Jiunn-Horng Kang
- Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, 252 Wuxing Str., Taipei 11031, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Str., Taipei 11031, Taiwan
- Research Center of Artificial Intelligence in Medicine, Taipei Medical University, 250 Wuxing Str., Taipei 11031, Taiwan
| |
Collapse
|
24
|
Yang H, Bullock WA, Myhal A, DeShield P, Duffy D, Main RP. Cancellous Bone May Have a Greater Adaptive Strain Threshold Than Cortical Bone. JBMR Plus 2021; 5:e10489. [PMID: 33977205 PMCID: PMC8101616 DOI: 10.1002/jbm4.10489] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/24/2021] [Accepted: 03/09/2021] [Indexed: 01/12/2023] Open
Abstract
Strain magnitude has a controlling influence on bone adaptive response. However, questions remain as to how and if cancellous and cortical bone tissues respond differently to varied strain magnitudes, particularly at a molecular level. The goal of this study was to characterize the time‐dependent gene expression, bone formation, and structural response of the cancellous and cortical bone of female C57Bl/6 mice to mechanical loading by applying varying load levels (low: −3.5 N; medium: −5.2 N; high: −7 N) to the skeleton using a mouse tibia loading model. The loading experiment showed that cortical bone mass at the tibial midshaft was significantly enhanced following all load levels examined and bone formation activities were particularly elevated at the medium and high loads applied. In contrast, for the proximal metaphyseal cancellous bone, only the high load led to significant increases in bone mass and bone formation indices. Similarly, expression of genes associated with inhibition of bone formation (e.g., Sost) was altered in the diaphyseal cortical bone at all load levels, but in the metaphyseal cortico‐cancellous bone only by the high load. Finite element analysis determined that the peak tensile or compressive strains that were osteogenic for the proximal cancellous bone under the high load were significantly greater than those that were osteogenic for the midshaft cortical tissues under the low load. These results suggest that the magnitude of the strain stimulus regulating structural, cellular, and molecular responses of bone to loading may be greater for the cancellous tissues than for the cortical tissues. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Haisheng Yang
- Department of Biomedical Engineering, Faculty of Environment and Life Beijing University of Technology Beijing China
| | | | - Alexandra Myhal
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences Purdue University West Lafayette IN USA
| | - Philip DeShield
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences Purdue University West Lafayette IN USA
| | - Daniel Duffy
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN USA
| | - Russell P Main
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences Purdue University West Lafayette IN USA.,Weldon School of Biomedical Engineering Purdue University West Lafayette IN USA
| |
Collapse
|
25
|
Carriero A, Javaheri B, Bassir Kazeruni N, Pitsillides AA, Shefelbine SJ. Age and Sex Differences in Load-Induced Tibial Cortical Bone Surface Strain Maps. JBMR Plus 2021; 5:e10467. [PMID: 33778328 PMCID: PMC7990149 DOI: 10.1002/jbm4.10467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/03/2021] [Indexed: 12/21/2022] Open
Abstract
Bone adapts its architecture to the applied load; however, it is still unclear how bone mechano‐adaptation is coordinated and why potential for adaptation adjusts during the life course. Previous animal models have suggested strain as the mechanical stimulus for bone adaptation, but yet it is unknown how mouse cortical bone load‐related strains vary with age and sex. In this study, full‐field strain maps (at 1 N increments up to 12 N) on the bone surface were measured in young, adult, and old (aged 10, 22 weeks, and 20 months, respectively), male and female C57BL/6J mice with load applied using a noninvasive murine tibial model. Strain maps indicate a nonuniform strain field across the tibial surface, with axial compressive loads resulting in tension on the medial side of the tibia because of its curved shape. The load‐induced surface strain patterns and magnitudes show sexually dimorphic changes with aging. A comparison of the average and peak tensile strains indicates that the magnitude of strain at a given load generally increases during maturation, with tibias in female mice having higher strains than in males. The data further reveal that postmaturation aging is linked to sexually dimorphic changes in average and maximum strains. The strain maps reported here allow for loading male and female C57BL/6J mouse legs in vivo at the observed ages to create similar increases in bone surface average or peak strain to more accurately explore bone mechano‐adaptation differences with age and sex. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alessandra Carriero
- Department of Biomedical Engineering The City College of New York New York NY USA
| | - Behzad Javaheri
- School of Mathematics, Computer Science and Engineering, City University of London London UK
| | | | - Andrew A Pitsillides
- Department of Comparative Biomedical Sciences Royal Veterinary College London UK
| | - Sandra J Shefelbine
- Department of Mechanical and Industrial Engineering and Department of Bioengineering Northeastern University Boston MA USA
| |
Collapse
|
26
|
Riquelme MA, Gu S, Hua R, Jiang JX. Mechanotransduction via the coordinated actions of integrins, PI3K signaling and Connexin hemichannels. Bone Res 2021; 9:8. [PMID: 33531460 PMCID: PMC7854719 DOI: 10.1038/s41413-020-00126-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/25/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Mechanical loading opens connexin 43 (Cx43) hemichannels (HCs), leading to the release of bone anabolic molecules, such as prostaglandins, from mechanosensitive osteocytes, which is essential for bone formation and remodeling. However, the mechanotransduction mechanism that activates HCs remains elusive. Here, we report a unique pathway by which mechanical signals are effectively transferred between integrin molecules located in different regions of the cell, resulting in HC activation. Both integrin α5 and αV were activated upon mechanical stimulation via either fluid dropping or flow shear stress (FSS). Inhibition of integrin αV activation or ablation of integrin α5 prevented HC opening on the cell body when dendrites were mechanically stimulated, suggesting mechanical transmission from the dendritic integrin αV to α5 in the cell body during HC activation. In addition, HC function was compromised in vivo, as determined by utilizing an antibody blocking αV activation and α5-deficient osteocyte-specific knockout mice. Furthermore, inhibition of integrin αV activation, but not that of α5, attenuated activation of the phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway upon mechanical loading, and the inhibition of PI3K/AKT activation blocked integrin α5 activation and HC opening. Moreover, HC opening was blocked only by an anti-integrin αV antibody at low but not high FSS levels, suggesting that dendritic αV is a more sensitive mechanosensor than α5 for activating HCs. Together, these results reveal a new molecular mechanism of mechanotransduction involving the coordinated actions of integrins and PI3K/AKT in osteocytic dendritic processes and cell bodies that leads to HC opening and the release of key bone anabolic factors.
Collapse
Affiliation(s)
- Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
27
|
Cortical bone adaptation to a moderate level of mechanical loading in male Sost deficient mice. Sci Rep 2020; 10:22299. [PMID: 33339872 PMCID: PMC7749116 DOI: 10.1038/s41598-020-79098-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/30/2020] [Indexed: 11/08/2022] Open
Abstract
Loss-of-function mutations in the Sost gene lead to high bone mass phenotypes. Pharmacological inhibition of Sost/sclerostin provides a new drug strategy for treating osteoporosis. Questions remain as to how physical activity may affect bone mass under sclerostin inhibition and if that effect differs between males and females. We previously observed in female Sost knockout (KO) mice an enhanced cortical bone formation response to a moderate level of applied loading (900 με at the tibial midshaft). The purpose of the present study was to examine cortical bone adaptation to the same strain level applied to male Sost KO mice. Strain-matched in vivo compressive loading was applied to the tibiae of 10-, 26- and 52-week-old male Sost KO and littermate control (LC) mice. The effect of tibial loading on bone (re)modeling was measured by microCT, 3D time-lapse in vivo morphometry, 2D histomorphometry and gene expression analyses. As expected, Sost deficiency led to high cortical bone mass in 10- and 26-week-old male mice as a result of increased bone formation. However, the enhanced bone formation associated with Sost deficiency did not appear to diminish with skeletal maturation. An increase in bone resorption was observed with skeletal maturation in male LC and Sost KO mice. Two weeks of in vivo loading (900 με at the tibial midshaft) induced only a mild anabolic response in 10- and 26-week-old male mice, independent of Sost deficiency. A decrease in the Wnt inhibitor Dkk1 expression was observed 3 h after loading in 52-week-old Sost KO and LC mice, and an increase in Lef1 expression was observed 8 h after loading in 10-week-old Sost KO mice. The current results suggest that long-term inhibition of sclerostin in male mice does not influence the adaptive response of cortical bone to moderate levels of loading. In contrast with our previous strain-matched study in females showing enhanced bone responses with Sost ablation, these results in males indicate that the influence of Sost deficiency on the cortical bone formation response to a moderate level of loading differs between males and females. Clinical studies examining antibodies to inhibit sclerostin may need to consider that the efficacy of additional physical activity regimens may be sex dependent.
Collapse
|
28
|
Pickering E, Silva MJ, Delisser P, Brodt MD, Gu Y, Pivonka P. Estimation of load conditions and strain distribution for in vivo murine tibia compression loading using experimentally informed finite element models. J Biomech 2020; 115:110140. [PMID: 33348259 DOI: 10.1016/j.jbiomech.2020.110140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022]
Abstract
The murine tibia compression model, is the gold standard for studying bone adaptation due to mechanical loading in vivo. Currently, a key limitation of the experimental protocol and associated finite element (FE) models is that the exact load transfer, and consequently the loading conditions on the tibial plateau, is unknown. Often in FE models, load is applied to the tibial plateau based on inferences from micro-computed tomography (μCT). Experimental models often use a single strain gauge to assess the three-dimensional (3D) loading state. However, a single strain gauge is insufficient to validate such FE models. To address this challenge, we develop an experimentally calibrated method for identifying the load application region on the tibial plateau based upon measurements from three strain gauges. To achieve this, axial compression was conducted on mouse tibiae (n=3), with strains gauges on three surfaces. FE simulations were performed to compute the strains at the gauge locations as a function of a variable load location. By minimising the error between experimental and FE strains, the precise load location was identified; this was found to vary between tibia specimens. It was further shown that commonly used FE loading conditions, found in literature, did not replicate the experimental strain distribution, highlighting the importance of load calibration. This work provides critical insights into how load is transferred to the tibial plateau. Importantly, this work develops an experimentally informed technique for loading the tibial plateau in FE models.
Collapse
Affiliation(s)
- Edmund Pickering
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, Saint Louis, MO, USA; Department of Biomedical Engineering, Washington University, Saint Louis, MO, USA
| | - Peter Delisser
- University of Bristol School of Veterinary Science, Bristol, UK; Veterinary Specialist Services, Brisbane, QLD, Australia
| | - Michael D Brodt
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, Saint Louis, MO, USA
| | - YuanTong Gu
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Peter Pivonka
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| |
Collapse
|
29
|
Yan C, Moshage SG, Kersh ME. Play During Growth: the Effect of Sports on Bone Adaptation. Curr Osteoporos Rep 2020; 18:684-695. [PMID: 33084999 DOI: 10.1007/s11914-020-00632-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW The development of exercise interventions for bone health requires an understanding of normative growth trends. Here, we summarize changes in bone during growth and the effect of participating in sports on structural and compositional measures in different bones in males and females. RECENT FINDINGS Growing females and males have similar normalized density and bone area fraction until age 16, after which males continue increasing at a faster rate than females. All metrics for both sexes tend to plateau or decline in the early 20s. Areal BMD measures indicate significant heterogeneity in adaptation to sport between regions of the body. High-resolution CT data indicate changes in structure are more readily apparent than changes in density. While adaptation to sport is spatially heterogeneous, participation in weight-bearing activities that involve dynamic muscle contractions tends to result in increased bone adaptation.
Collapse
Affiliation(s)
- Chenxi Yan
- Tissue Biomechanics Lab, Department of Mechanical Science and Engineering, Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Sara G Moshage
- Tissue Biomechanics Lab, Department of Mechanical Science and Engineering, Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Mariana E Kersh
- Tissue Biomechanics Lab, Department of Mechanical Science and Engineering, Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
30
|
Chermside-Scabbo CJ, Harris TL, Brodt MD, Braenne I, Zhang B, Farber CR, Silva MJ. Old Mice Have Less Transcriptional Activation But Similar Periosteal Cell Proliferation Compared to Young-Adult Mice in Response to in vivo Mechanical Loading. J Bone Miner Res 2020; 35:1751-1764. [PMID: 32311160 PMCID: PMC7486279 DOI: 10.1002/jbmr.4031] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Mechanical loading is a potent strategy to induce bone formation, but with aging, the bone formation response to the same mechanical stimulus diminishes. Our main objectives were to (i) discover the potential transcriptional differences and (ii) compare the periosteal cell proliferation between tibias of young-adult and old mice in response to strain-matched mechanical loading. First, to discover potential age-related transcriptional differences, we performed RNA sequencing (RNA-seq) to compare the loading responses between tibias of young-adult (5-month) and old (22-month) C57BL/6N female mice following 1, 3, or 5 days of axial loading (loaded versus non-loaded). Compared to young-adult mice, old mice had less transcriptional activation following loading at each time point, as measured by the number of differentially expressed genes (DEGs) and the fold-changes of the DEGs. Old mice engaged fewer pathways and gene ontology (GO) processes, showing less activation of processes related to proliferation and differentiation. In tibias of young-adult mice, we observed prominent Wnt signaling, extracellular matrix (ECM), and neuronal responses, which were diminished with aging. Additionally, we identified several targets that may be effective in restoring the mechanoresponsiveness of aged bone, including nerve growth factor (NGF), Notum, prostaglandin signaling, Nell-1, and the AP-1 family. Second, to directly test the extent to which periosteal cell proliferation was diminished in old mice, we used bromodeoxyuridine (BrdU) in a separate cohort of mice to label cells that divided during the 5-day loading interval. Young-adult and old mice had an average of 15.5 and 16.7 BrdU+ surface cells/mm, respectively, suggesting that impaired proliferation in the first 5 days of loading does not explain the diminished bone formation response with aging. We conclude that old mice have diminished transcriptional activation following mechanical loading, but periosteal proliferation in the first 5 days of loading does not differ between tibias of young-adult and old mice. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christopher J Chermside-Scabbo
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Medical Scientist Training Program, Washington University School of Medicine, Washington University, St. Louis, MO, USA
| | - Taylor L Harris
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Michael D Brodt
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Ingrid Braenne
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Department of Developmental Biology, Washington University, St. Louis, MO, USA
| | - Charles R Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Matthew J Silva
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| |
Collapse
|
31
|
Scheuren AC, D'Hulst G, Kuhn GA, Masschelein E, Wehrle E, De Bock K, Müller R. Hallmarks of frailty and osteosarcopenia in prematurely aged PolgA (D257A/D257A) mice. J Cachexia Sarcopenia Muscle 2020; 11:1121-1140. [PMID: 32596975 PMCID: PMC7432580 DOI: 10.1002/jcsm.12588] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Frailty is a geriatric syndrome characterized by increased susceptibility to adverse health outcomes. One major determinant thereof is the gradual weakening of the musculoskeletal system and the associated osteosarcopenia. To improve our understanding of the underlying pathophysiology and, more importantly, to test potential interventions aimed at counteracting frailty, suitable animal models are needed. METHODS To evaluate the relevance of prematurely aged PolgA(D257A/D257A) mice as a model for frailty and osteosarcopenia, we quantified the clinical mouse frailty index in PolgA(D257A/D257A) and wild-type littermates (PolgA(+/+) , WT) with age and concertedly assessed the quantity and quality of bone and muscle tissue. Lastly, the anabolic responsiveness of skeletal muscle, muscle progenitors, and bone was assessed. RESULTS PolgA(D257A/D257A) accumulated health deficits at a higher rate compared with WT, resulting in a higher frailty index at 40 and 46 weeks of age (+166%, +278%, P < 0.0001), respectively, with no differences between genotypes at 34 weeks. Concomitantly, PolgA(D257A/D257A) displayed progressive musculoskeletal deterioration such as reduced bone and muscle mass as well as impaired functionality thereof. In addition to lower muscle weights (-14%, P < 0.05, -23%, P < 0.0001) and fibre area (-20%, P < 0.05, -22%, P < 0.0001) at 40 and 46 weeks, respectively, PolgA(D257A/D257A) showed impairments in grip strength and concentric muscle forces (P < 0.05). PolgA(D257A/D257A) mutation altered the acute response to various anabolic stimuli in skeletal muscle and muscle progenitors. While PolgA(D257A/D257A) muscles were hypersensitive to eccentric contractions as well as leucine administration, shown by larger downstream signalling response of the mechanistic target of rapamycin complex 1, myogenic progenitors cultured in vitro showed severe anabolic resistance to leucine and robust impairments in cell proliferation. Longitudinal micro-computed tomography analysis of the sixth caudal vertebrae showed that PolgA(D257A/D257A) had lower bone morphometric parameters (e.g. bone volume fraction, trabecular, and cortical thickness, P < 0.05) as well as reduced remodelling activities (e.g. bone formation and resorption rate, P < 0.05) compared with WT. When subjected to 4 weeks of cyclic loading, young but not aged PolgA(D257A/D257A) caudal vertebrae showed load-induced bone adaptation, suggesting reduced mechanosensitivity with age. CONCLUSIONS PolgA(D257A/D257A) mutation leads to hallmarks of age-related frailty and osteosarcopenia and provides a powerful model to better understand the relationship between frailty and the aging musculoskeletal system.
Collapse
Affiliation(s)
| | - Gommaar D'Hulst
- Laboratory of Exercise and HealthETH ZurichZurichSwitzerland
| | | | - Evi Masschelein
- Laboratory of Exercise and HealthETH ZurichZurichSwitzerland
| | - Esther Wehrle
- Institute for BiomechanicsETH ZurichZurichSwitzerland
| | - Katrien De Bock
- Laboratory of Exercise and HealthETH ZurichZurichSwitzerland
| | - Ralph Müller
- Institute for BiomechanicsETH ZurichZurichSwitzerland
| |
Collapse
|
32
|
Rooney AM, Bostrom MPG, Dempster DW, Nieves JW, Zhou H, Cosman F. Loading modality and age influence teriparatide-induced bone formation in the human femoral neck. Bone 2020; 136:115373. [PMID: 32330694 PMCID: PMC7263665 DOI: 10.1016/j.bone.2020.115373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/01/2020] [Accepted: 04/18/2020] [Indexed: 12/14/2022]
Abstract
Teriparatide (TPTD) reduces risk of both vertebral and nonvertebral fracture, but increases bone mineral density (BMD) much more at the spine than the hip. TPTD and mechanical loading may have a synergistic anabolic effect on BMD, which may help explain these site-specific differences. Under normal daily activity, the femoral neck (FN) is under bending, placing one side under tension and the other under compression. We sought to further understand the relationship between mechanical loading and TPTD at the hip by investigating the effect of tensile versus compressive loading on TPTD stimulated bone formation indices in the human FN. Thirty-eight patients receiving total hip replacements for osteoarthritis were randomized to receive placebo (PBO) or TPTD for a mean treatment duration of 6 weeks prior to surgery, and double tetracycline labeling was administered to allow assessment of bone formation. The FN was harvested during surgery and analyzed for dynamic bone formation indices in the compressive and tensile regions of the endocortical and periosteal envelopes. Regression models relating outcome measures to patient characteristics including sex, age, body weight, and FN geometry were also analyzed. Overall, bone formation was higher with TPTD versus placebo on the endocortical surface, but not the periosteal surface. The level of bone formation in both TPTD and placebo groups was greater on the tensile endocortical surface and the compressive periosteal surface. There was a trend toward decreased endocortical eroded surface with TPTD in the compressive but not the tensile region. Patient age and sex explained the greatest variability in endocortical bone formation, and patient body mass and sex explained the greatest variability in periosteal bone formation. Our data represent the first dynamic comparison of teriparatide treatment under two loading modalities in human FN samples. Future work could determine whether specific hip loading intervention could amplify the benefits of teriparatide on the hip in clinical settings.
Collapse
Affiliation(s)
- Amanda M Rooney
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, Ithaca, NY 14853, USA.
| | - Mathias P G Bostrom
- Research Division, Hospital for Special Surgery, 515 East 71st St., New York, NY 10021, USA.
| | - David W Dempster
- Regional Bone Center, Helen Hayes Hospital, 55 N Route 9W, West Haverstraw, NY 10993, USA; Department of Pathology, Columbia University, 630 West 168th St., New York, NY 10025, USA
| | - Jeri W Nieves
- Research Division, Hospital for Special Surgery, 515 East 71st St., New York, NY 10021, USA; Regional Bone Center, Helen Hayes Hospital, 55 N Route 9W, West Haverstraw, NY 10993, USA; Department of Epidemiology, Columbia University, 722 West 168th St., New York, NY 10032, USA.
| | - Hua Zhou
- Regional Bone Center, Helen Hayes Hospital, 55 N Route 9W, West Haverstraw, NY 10993, USA.
| | - Felicia Cosman
- Department of Medicine, Columbia University, 622 West 168th St., New York, NY 10032, USA.
| |
Collapse
|
33
|
Qin L, Liu W, Cao H, Xiao G. Molecular mechanosensors in osteocytes. Bone Res 2020; 8:23. [PMID: 32550039 PMCID: PMC7280204 DOI: 10.1038/s41413-020-0099-y] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/07/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Osteocytes, the most abundant and long-lived cells in bone, are the master regulators of bone remodeling. In addition to their functions in endocrine regulation and calcium and phosphate metabolism, osteocytes are the major responsive cells in force adaptation due to mechanical stimulation. Mechanically induced bone formation and adaptation, disuse-induced bone loss and skeletal fragility are mediated by osteocytes, which sense local mechanical cues and respond to these cues in both direct and indirect ways. The mechanotransduction process in osteocytes is a complex but exquisite regulatory process between cells and their environment, between neighboring cells, and between different functional mechanosensors in individual cells. Over the past two decades, great efforts have focused on finding various mechanosensors in osteocytes that transmit extracellular mechanical signals into osteocytes and regulate responsive gene expression. The osteocyte cytoskeleton, dendritic processes, Integrin-based focal adhesions, connexin-based intercellular junctions, primary cilium, ion channels, and extracellular matrix are the major mechanosensors in osteocytes reported so far with evidence from both in vitro and in vitro studies. This review aims to give a systematic introduction to osteocyte mechanobiology, provide details of osteocyte mechanosensors, and discuss the roles of osteocyte mechanosensitive signaling pathways in the regulation of bone homeostasis.
Collapse
Affiliation(s)
- Lei Qin
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Wen Liu
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Huiling Cao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Guozhi Xiao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| |
Collapse
|
34
|
Adebayo OO, Holyoak DT, van der Meulen MCH. Mechanobiological Mechanisms of Load-Induced Osteoarthritis in the Mouse Knee. J Biomech Eng 2020; 141:2736041. [PMID: 31209459 DOI: 10.1115/1.4043970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects millions of people worldwide, yet its disease mechanism is not clearly understood. Animal models have been established to study disease progression by initiating OA through modified joint mechanics or altered biological activity within the joint. However, animal models often do not have the capability to directly relate the mechanical environment to joint damage. This review focuses on a novel in vivo approach based on controlled, cyclic tibial compression to induce OA in the mouse knee. First, we discuss the development of the load-induced OA model, its different loading configurations, and other techniques used by research laboratories around the world. Next, we review the lessons learned regarding the mechanobiological mechanisms of load-induced OA and relate these findings to the current understanding of the disease. Then, we discuss the role of specific genetic and cellular pathways involved in load-induced OA progression and the contribution of altered tissue properties to the joint response to mechanical loading. Finally, we propose using this approach to test the therapeutic efficacy of novel treatment strategies for OA. Ultimately, elucidating the mechanobiological mechanisms of load-induced OA will aid in developing targeted treatments for this disabling disease.
Collapse
Affiliation(s)
| | - Derek T Holyoak
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering, Cornell University, 113 Weill Hall, Ithaca, NY 14853.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853.,Research Division, Hospital for Special Surgery, New York, NY 10021 e-mail:
| |
Collapse
|
35
|
Asgharzadeh P, Röhrle O, Willie BM, Birkhold AI. Decoding rejuvenating effects of mechanical loading on skeletal aging using in vivo μCT imaging and deep learning. Acta Biomater 2020; 106:193-207. [PMID: 32058080 DOI: 10.1016/j.actbio.2020.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 12/21/2022]
Abstract
Throughout the process of aging, dynamic changes of bone material, micro- and macro-architecture result in a loss of strength and therefore in an increased likelihood of fragility fractures. To date, precise contributions of age-related changes in bone (re)modeling and (de)mineralization dynamics to this fragility increase are not completely understood. Here, we present an image-based deep learning approach to quantitatively describe the effects of short-term aging and adaptive response to cyclic loading applied to proximal mouse tibiae and fibulae. Our approach allowed us to perform an end-to-end age prediction based on μCT imaging to determine the dynamic biological process of aging during a two week period, therefore permitting short-term bone aging analysis with 95% accuracy in predicting time points. In a second application, our deep learning analysis reveals that two weeks of in vivo mechanical loading are associated with an underlying rejuvenating effect of 5 days. Additionally, by quantitatively analyzing the learning process, we could, for the first time, identify the localization of the age-relevant encoded information and demonstrate 89% load-induced similarity of these locations in the loaded tibia with younger control bones. These data therefore suggest that our method enables identifying a general prognostic phenotype of a certain skeletal age as well as a temporal and localized loading-treatment effect on this apparent skeletal age for the studied mouse tibia and fibula. Future translational applications of this method may provide an improved decision-support method for osteoporosis treatment at relatively low cost. STATEMENT OF SIGNIFICANCE: Bone is a highly complex and dynamic structure that undergoes changes during the course of aging as well as in response to external stimuli, such as loading. Automatic assessment of "age" and "state" of the bone may lead to early prognosis of deceases such as osteoporosis and enables evaluating the effects of certain treatments. Here, we present an artificial intelligence-based method capable of automatically predicting the skeletal age from μCT images with 95% accuracy. Additionally, we utilize it to demonstrate the rejuvenation effects of in-vivo loading treatment on bones. We further, for the first time, break down aging-related local changes in bone by quantitatively analyzing "what the age assessment model has learned" and use this information to investigate the structural details of rejuvenation process.
Collapse
Affiliation(s)
- Pouyan Asgharzadeh
- Institute for Modelling and Simulation of Biomechanical Systems, University of Stuttgart, Stuttgart, Germany; Stuttgart Center for Simulation Science (SC SimTech), Stuttgart, Germany. http://bit.ly/2Tqx_PA
| | - Oliver Röhrle
- Institute for Modelling and Simulation of Biomechanical Systems, University of Stuttgart, Stuttgart, Germany; Stuttgart Center for Simulation Science (SC SimTech), Stuttgart, Germany
| | - Bettina M Willie
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Canada
| | - Annette I Birkhold
- Institute for Modelling and Simulation of Biomechanical Systems, University of Stuttgart, Stuttgart, Germany; Stuttgart Center for Simulation Science (SC SimTech), Stuttgart, Germany
| |
Collapse
|
36
|
Main RP, Shefelbine SJ, Meakin LB, Silva MJ, van der Meulen MC, Willie BM. Murine Axial Compression Tibial Loading Model to Study Bone Mechanobiology: Implementing the Model and Reporting Results. J Orthop Res 2020; 38:233-252. [PMID: 31508836 PMCID: PMC9344861 DOI: 10.1002/jor.24466] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/23/2019] [Indexed: 02/04/2023]
Abstract
In vivo, tibial loading in mice is increasingly used to study bone adaptation and mechanotransduction. To achieve standardized and defined experimental conditions, loading parameters and animal-related factors must be considered when performing in vivo loading studies. In this review, we discuss these loading and animal-related experimental conditions, present methods to assess bone adaptation, and suggest reporting guidelines. This review originated from presentations by each of the authors at the workshop "Developing Best Practices for Mouse Models of In Vivo Loading" during the Preclinical Models Section at the Orthopaedic Research Society Annual Meeting, San Diego, CA, March 2017. Following the meeting, the authors engaged in detailed discussions with consideration of relevant literature. The guidelines and recommendations in this review are provided to help researchers perform in vivo loading experiments in mice, and thus further our knowledge of bone adaptation and the mechanisms involved in mechanotransduction. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:233-252, 2020.
Collapse
Affiliation(s)
- Russell P. Main
- Department of Basic Medical Sciences and Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA,Corresponding author: Russell Main ()
| | - Sandra J. Shefelbine
- Department of Bioengineering, Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, USA
| | - Lee B. Meakin
- Bristol Veterinary School, University of Bristol, Langford, Bristol BS40 5DU, UK
| | - Matthew J. Silva
- Departments of Orthopaedic Surgery and Biomedical Engineering, Musculoskeletal Research Center, Washington University, Saint Louis, MO, USA
| | - Marjolein C.H van der Meulen
- Meinig School of Biomedical Engineering and Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Bettina M. Willie
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| |
Collapse
|
37
|
Morrell AE, Robinson ST, Silva MJ, Guo XE. Mechanosensitive Ca 2+ signaling and coordination is diminished in osteocytes of aged mice during ex vivo tibial loading. Connect Tissue Res 2020; 61:389-398. [PMID: 31931640 PMCID: PMC7785095 DOI: 10.1080/03008207.2020.1712377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose: The osteocyte is considered the major mechanosensor in bone, capable of detecting forces at a cellular level to coordinate bone formation and resorption. The pathology of age-related bone loss, a hallmark of osteoporosis, is attributed in part to impaired osteocyte mechanosensing. However, real-time evidence of the effect of aging on osteocyte responses to mechanical load is lacking. Intracellular calcium (Ca2+) oscillations have been characterized as an early mechanosensitive response in osteocytes in systems of multiple scales and thus can serve as a real-time measure of osteocyte mechanosensitivity. Our objective was to utilize an ex vivo model to investigate potentially altered mechanosensing in the osteocyte network with aging.Methods: Tibiae were explanted from young-adult (5 mo) and aged (22 mo) female mice and incubated with Fluo-8 AM to visualize osteocyte intracellular Ca2+. Whole tibiae were cyclically loaded while in situ osteocyte Ca2+ dynamics were simultaneously imaged with confocal microscopy. Responsive osteocyte percentage and Ca2+ peak characteristics were quantified, as well as signaling synchrony between paired cells in the field of view.Results: Fewer osteocytes responded to mechanical loading in aged mice compared to young-adult and did so in a delayed manner. Osteocytes from aged mice also lacked the well-correlated relationship between Ca2+ signaling synchrony and cell-cell distance exhibited by young-adult osteocytes.Conclusions: We have demonstrated, for the first time, real-time evidence of the diminished mechanosensing and lack of signaling coordination in aged osteocyte networks in tibial explants, which may contribute to pathology of age-induced bone loss.
Collapse
Affiliation(s)
- Andrea E. Morrell
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Samuel T. Robinson
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, USA,Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - X. Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA,Corresponding author: X. Edward Guo, PhD, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904,1210 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
38
|
Curtis KJ, Oberman AG, Niebur GL. Effects of mechanobiological signaling in bone marrow on skeletal health. Ann N Y Acad Sci 2019; 1460:11-24. [PMID: 31508828 DOI: 10.1111/nyas.14232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 01/27/2023]
Abstract
Bone marrow is a cellular tissue that forms within the pore space and hollow diaphysis of bones. As a tissue, its primary function is to support the hematopoietic progenitor cells that maintain the populations of both erythroid and myeloid lineage cells in the bone marrow, making it an essential element of normal mammalian physiology. However, bone's primary function is load bearing, and deformations induced by external forces are transmitted to the encapsulated marrow. Understanding the effects of these mechanical inputs on marrow function and adaptation requires knowledge of the material behavior of the marrow at multiple scales, the loads that are applied, and the mechanobiology of the cells. This paper reviews the current state of knowledge of each of these factors. Characterization of the marrow mechanical environment and its role in skeletal health and other marrow functions remains incomplete, but research on the topic is increasing, driven by interest in skeletal adaptation and the mechanobiology of cancer metastasis.
Collapse
Affiliation(s)
- Kimberly J Curtis
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana.,Advanced Diagnostics and Therapeutics Initiative, University of Notre Dame, Notre Dame, Indiana
| | - Alyssa G Oberman
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana
| | - Glen L Niebur
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
39
|
Zimmermann EA, Riedel C, Schmidt FN, Stockhausen KE, Chushkin Y, Schaible E, Gludovatz B, Vettorazzi E, Zontone F, Püschel K, Amling M, Ritchie RO, Busse B. Mechanical Competence and Bone Quality Develop During Skeletal Growth. J Bone Miner Res 2019; 34:1461-1472. [PMID: 30913317 DOI: 10.1002/jbmr.3730] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 02/01/2023]
Abstract
Bone fracture risk is influenced by bone quality, which encompasses bone's composition as well as its multiscale organization and architecture. Aging and disease deteriorate bone quality, leading to reduced mechanical properties and higher fracture incidence. Largely unexplored is how bone quality and mechanical competence progress during longitudinal bone growth. Human femoral cortical bone was acquired from fetal (n = 1), infantile (n = 3), and 2- to 14-year-old cases (n = 4) at the mid-diaphysis. Bone quality was assessed in terms of bone structure, osteocyte characteristics, mineralization, and collagen orientation. The mechanical properties were investigated by measuring tensile deformation at multiple length scales via synchrotron X-ray diffraction. We find dramatic differences in mechanical resistance with age. Specifically, cortical bone in 2- to 14-year-old cases exhibits a 160% greater stiffness and 83% higher strength than fetal/infantile cases. The higher mechanical resistance of the 2- to 14-year-old cases is associated with advantageous bone quality, specifically higher bone volume fraction, better micronscale organization (woven versus lamellar), and higher mean mineralization compared with fetal/infantile cases. Our study reveals that bone quality is superior after remodeling/modeling processes convert the primary woven bone structure to lamellar bone. In this cohort of female children, the microstructural differences at the femoral diaphysis were apparent between the 1- to 2-year-old cases. Indeed, the lamellar bone in 2- to 14-year-old cases had a superior structural organization (collagen and osteocyte characteristics) and composition for resisting deformation and fracture than fetal/infantile bone. Mechanistically, the changes in bone quality during longitudinal bone growth lead to higher fracture resistance because collagen fibrils are better aligned to resist tensile forces, while elevated mean mineralization reinforces the collagen scaffold. Thus, our results reveal inherent weaknesses of the fetal/infantile skeleton signifying its inferior bone quality. These results have implications for pediatric fracture risk, as bone produced at ossification centers during children's longitudinal bone growth could display similarly weak points. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Christoph Riedel
- Department of Osteology and Biomechanics, University Medical Center, Hamburg, Germany
| | - Felix N Schmidt
- Department of Osteology and Biomechanics, University Medical Center, Hamburg, Germany
| | - Kilian E Stockhausen
- Department of Osteology and Biomechanics, University Medical Center, Hamburg, Germany
| | - Yuriy Chushkin
- Beamline ID 10, European Synchrotron Radiation Facility, Grenoble, France
| | - Eric Schaible
- Experimental Systems Group, Advanced Light Source, Berkeley, CA, USA
| | - Bernd Gludovatz
- School of Mechanical and Manufacturing Engineering, UNSW Sydney, NSW, Australia
| | - Eik Vettorazzi
- Department of Medical Biometry and Epidemiology, University Medical Center, Hamburg, Germany
| | - Federico Zontone
- Beamline ID 10, European Synchrotron Radiation Facility, Grenoble, France
| | - Klaus Püschel
- Department of Forensic Medicine, University Medical Center, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center, Hamburg, Germany
| | - Robert O Ritchie
- Department of Materials Science and Engineering, University of California, Berkeley, CA, USA.,Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center, Hamburg, Germany
| |
Collapse
|
40
|
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: 1.8] [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
|
41
|
Suzuki H, Tatei K, Ohshima N, Sato S, Izumi T. Regulation of MC3T3-E1 differentiation by actin cytoskeleton through lipid mediators reflecting the cell differentiation stage. Biochem Biophys Res Commun 2019; 514:393-400. [PMID: 31047639 DOI: 10.1016/j.bbrc.2019.04.093] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/12/2019] [Indexed: 12/20/2022]
Abstract
Actin cytoskeleton is reported to be related in various functions of osteoblast, bone-forming cell. However the function of actin cytoskeleton in osteoblasts is not fully understood, since bone formation is derived from extracellular interactions of functional proteins produced from osteoblasts, including osteocalcin (Ocn), and it is a result of closely and complex organized sequence of biochemical events. In this study, we showed that actin cytoskeleton of MC3T3-E1 cells functioned in recognition of cell condition and regulation of extracellular matrix mineralization, bone formation. Maturation of MC3T3-E1 cells by 14 days of culture reduced F-actin filaments, while induced expression of Ocn mRNA known as late stage differentiation marker and matrix mineralization, terminal stage of cell differentiation. The disruption of actin cytoskeleton with Cyto D in immature MC3T3-E1 cells significantly increased expression of Ocn mRNA in 24 h. Both PTX-induced inhibition of signal transduction through GPCRs and celecoxib-induced suppression of lipid mediators in immature MC3T3-E1 cells reduced actin filaments and suppressed matrix mineralization. Furthermore, addition of lipid mediators extracted from culture mediums of differentiated MC3T3-E1 cells by Bligh-Dyer method induced actin cytoskeleton reorganization and matrix mineralization change in MC3T3-E1 cells. Taken together, our data suggest that actin cytoskeleton of MC3T3-E1 cells regulates activation of developmental pathway reflecting cell differentiation stages through lipid mediators. The function we identified is important for bone formation tightly regulated by mechanical stress, since actin cytoskeleton is also known as a mechanosensor of osteoblasts.
Collapse
Affiliation(s)
- Hiraku Suzuki
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, Gunma, 371-8511, Japan.
| | - Kazuaki Tatei
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, Gunma, 371-8511, Japan
| | - Noriyasu Ohshima
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, Gunma, 371-8511, Japan
| | - Seiichi Sato
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
| | - Takashi Izumi
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
42
|
Yang H, Xu X, Bullock W, Main RP. Adaptive changes in micromechanical environments of cancellous and cortical bone in response to in vivo loading and disuse. J Biomech 2019; 89:85-94. [PMID: 31047696 DOI: 10.1016/j.jbiomech.2019.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 04/12/2019] [Accepted: 04/12/2019] [Indexed: 10/27/2022]
Abstract
The skeleton accommodates changes in mechanical environments by increasing bone mass under increased loads and decreasing bone mass under disuse. However, little is known about the adaptive changes in micromechanical behavior of cancellous and cortical tissues resulting from loading or disuse. To address this issue, in vivo tibial loading and hindlimb unloading experiments were conducted on 16-week-old female C57BL/6J mice. Changes in bone mass and tissue-level strains in the metaphyseal cancellous and midshaft cortical bone of the tibiae, resulting from loading or unloading, were determined using microCT and finite element (FE) analysis, respectively. We found that loading- and unloading-induced changes in bone mass were more pronounced in the cancellous than cortical bone. Simulated FE-loading showed that a greater proportion of elements experienced relatively lower longitudinal strains following load-induced bone adaptation, while the opposite was true in the disuse model. While the magnitudes of maximum or minimum principal strains in the metaphyseal cancellous and midshaft cortical bone were not affected by loading, strains oriented with the long axis were reduced in the load-adapted tibia suggesting that loading-induced micromechanical benefits were aligned primarily in the loading direction. Regression analyses demonstrated that bone mass was a good predictor of bone tissue strains for the cortical bone but not for the cancellous bone, which has complex microarchitecture and spatially-variant strain environments. In summary, loading-induced micromechanical benefits for cancellous and cortical tissues are received primarily in the direction of force application and cancellous bone mass may not be related to the micromechanics of cancellous bone.
Collapse
Affiliation(s)
- Haisheng Yang
- Department of Biomedical Engineering, School of Life Science and Bioengineering, Beijing University of Technology, Intelligent Physiological Measurement and Clinical Translation Beijing International Base for Scientific and Technological Cooperation, Beijing, China.
| | - Xiaoyu Xu
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, IN, USA; Weldon School of Biomedical Engineering, Purdue University, IN, USA.
| | | | - Russell P Main
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, IN, USA; Weldon School of Biomedical Engineering, Purdue University, IN, USA.
| |
Collapse
|
43
|
Piet J, Hu D, Baron R, Shefelbine SJ. Bone adaptation compensates resorption when sciatic neurectomy is followed by low magnitude induced loading. Bone 2019; 120:487-494. [PMID: 30586636 DOI: 10.1016/j.bone.2018.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 01/02/2023]
Abstract
The uniaxial tibial loading model is commonly used to promote bone formation through mechanoadaptation in mice. Sciatic neurectomy on the other hand recruits osteoclasts, which results in bone loss. Previous studies have shown that combining sciatic neurectomy with high magnitude loading increases the amount of bone formed. Here we determine whether low-intensity loading (low magnitude and few cycles) is sufficient to maintain bone mass after sciatic neurectomy, either by promoting bone formation (balance between concurrent resorption and formation), or by preventing bone resorption altogether. We examined bone adaptation in 4 groups of female C57BL/6J mice, 19-22 weeks old: (1) sham surgery +10 N loading; (2) sham surgery +5 N loading; (3) sciatic neurectomy; (4) sciatic neurectomy +5 N loading. Left legs were kept intact as internal controls. We examined changes in bone cross sectional properties and marrow area with micro-CT images, and histomorphometric measures with histological sections at the midpoint between tibiofibular junctions. Loading at 10 N caused a significant increase in the amount of bone, but bone formation after 5 N of loading was not detectable in micro-CT images. There was significant bone loss in mice with sciatic neurectomy alone, but when combined with loading there was no significant bone loss. Histomorphometric analyses showed that loading at 5 N augmented bone formation periosteally on the lateral and posterior-medial surfaces, and reduced the number of endosteal osteoclasts on the posterior-medial surface compared to the contralateral leg. Combining sciatic neurectomy and loading at 5 N promoted faster mineral apposition on the periosteal lateral surface and augmented bone resorption on the endosteal posterior surface compared to the contralateral leg. These data demonstrate that low-intensity loading is sufficient to maintain bone mass after sciatic neurectomy, both by preventing recruitment of osteoclasts on the endosteal surface and by compensating endosteal resorption caused by disuse with periosteal formation promoted by loading. This has implications for the loading required to maintain bone mass after injury or prolonged bedrest.
Collapse
Affiliation(s)
- Judith Piet
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Dorothy Hu
- Department of Medicine, Harvard Medical School, Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Roland Baron
- Department of Medicine, Harvard Medical School, Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA; Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Berman AG, Hinton MJ, Wallace JM. Treadmill running and targeted tibial loading differentially improve bone mass in mice. Bone Rep 2019; 10:100195. [PMID: 30701187 PMCID: PMC6348199 DOI: 10.1016/j.bonr.2019.100195] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/28/2018] [Accepted: 01/14/2019] [Indexed: 01/28/2023] Open
Abstract
Treadmill running and tibial loading are two common modalities used to assess the role of mechanical stimulation on the skeleton preclinically. The primary advantage of treadmill running is its physiological relevance. However, the applied load is complex and multiaxial, with observed results influenced by cardiovascular and musculoskeletal effects. In contrast, with tibial loading, a direct uniaxial load is applied to a single bone, providing the advantage of greater control but with less physiological relevance. Despite the importance and wide-spread use of both modalities, direct comparisons are lacking. In this study, we compared effects of targeted tibial loading, treadmill running, and their combination on cancellous and cortical architecture in a murine model. We show that tibial loading and treadmill running differentially improve bone mass, with tibial loading resulting in thicker trabeculae and increased cortical mass, and exercise resulting in greater number of trabeculae and no cortical mass-based effects. Combination of the modalities resulted in an additive response. These data suggest that tibial loading and exercise may improve mass differentially. Tibial loading increased trabecular thickness while exercise increased number. Combined effects of loading and exercise were additive in cancellous bone. In cortical bone, loading increased cross-sectional area. No mass-based effects were noted due to exercise.
Collapse
Affiliation(s)
- Alycia G Berman
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Madicyn J Hinton
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
45
|
Hemmatian H, Jalali R, Semeins CM, Hogervorst JMA, van Lenthe GH, Klein-Nulend J, Bakker AD. Mechanical Loading Differentially Affects Osteocytes in Fibulae from Lactating Mice Compared to Osteocytes in Virgin Mice: Possible Role for Lacuna Size. Calcif Tissue Int 2018; 103:675-685. [PMID: 30109376 PMCID: PMC6208961 DOI: 10.1007/s00223-018-0463-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/30/2018] [Indexed: 12/31/2022]
Abstract
Hormonal changes during lactation are associated with profound changes in bone cell biology, such as osteocytic osteolysis, resulting in larger lacunae. Larger lacuna shape theoretically enhances the transmission of mechanical signals to osteocytes. We aimed to provide experimental evidence supporting this theory by comparing the mechanoresponse of osteocytes in the bone of lactating mice, which have enlarged lacunae due to osteocytic osteolysis, with the response of osteocytes in bone from age-matched virgin mice. The osteocyte mechanoresponse was measured in excised fibulae that were cultured in hormone-free medium for 24 h and cyclically loaded for 10 min (sinusoidal compressive load, 3000 µε, 5 Hz) by quantifying loading-related changes in Sost mRNA expression (qPCR) and sclerostin and β-catenin protein expression (immunohistochemistry). Loading decreased Sost expression by ~ threefold in fibulae of lactating mice. The loading-induced decrease in sclerostin protein expression by osteocytes was larger in lactating mice (55% decrease ± 14 (± SD), n = 8) than virgin mice (33% decrease ± 15, n = 7). Mechanical loading upregulated β-catenin expression in osteocytes in lactating mice by 3.5-fold (± 0.2, n = 6) which is significantly (p < 0.01) higher than the 1.6-fold increase in β-catenin expression by osteocytes in fibulae from virgin mice (± 0.12, n = 4). These results suggest that osteocytes in fibulae from lactating mice with large lacunae may respond stronger to mechanical loading than those from virgin mice. This could indicate that osteocytes residing in larger lacuna show a stronger response to mechanical loading.
Collapse
Affiliation(s)
- Haniyeh Hemmatian
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Rozita Jalali
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Cornelis M Semeins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Jolanda M A Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - G Harry van Lenthe
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands.
| | - Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Srinivasan S, Balsiger D, Huber P, Ausk BJ, Bain SD, Gardiner EM, Gross TS. Static Preload Inhibits Loading-Induced Bone Formation. JBMR Plus 2018; 3:e10087. [PMID: 31131340 DOI: 10.1002/jbm4.10087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
Nearly all exogenous loading models of bone adaptation apply dynamic loading superimposed upon a time invariant static preload (SPL) in order to ensure stable, reproducible loading of bone. Given that SPL may alter aspects of bone mechanotransduction (eg, interstitial fluid flow), we hypothesized that SPL inhibits bone formation induced by dynamic loading. As a first test of this hypothesis, we utilized a newly developed device that enables stable dynamic loading of the murine tibia with SPLs ≥ -0.01 N. We subjected the right tibias of BALB/c mice (4-month-old females) to dynamic loading (-3.8 N, 1 Hz, 50 cycles/day, 10 s rest) superimposed upon one of three SPLs: -1.5 N, -0.5 N, or -0.03 N. Mice underwent exogenous loading 3 days/week for 3 weeks. Metaphyseal trabecular bone adaptation (μCT) and midshaft cortical bone formation (dynamic histomorphometry) were assessed following euthanasia (day 22). Ipsilateral tibias of mice loaded with a -1.5-N SPL demonstrated significantly less trabecular bone volume/total volume (BV/TV) than contralateral tibias (-12.9%). In contrast, the same dynamic loading superimposed on a -0.03-N SPL significantly elevated BV/TV versus contralateral tibias (12.3%) and versus the ipsilateral tibias of the other SPL groups (-0.5 N: 46.3%, -1.5 N: 37.2%). At the midshaft, the periosteal bone formation rate (p.BFR) induced when dynamic loading was superimposed on -1.5-N and -0.5-N SPLs was significantly amplified in the -0.03-N SPL group (>200%). These data demonstrate that bone anabolism induced by dynamic loading is markedly inhibited by SPL magnitudes commonly implemented in the literature (ie, -0.5 N, -1.5 N). The inhibitory impact of SPL has not been recognized in bone adaptation models and, as such, SPLs have been neither universally reported nor standardized. Our study therefore identifies a previously unrecognized, potent inhibitor of mechanoresponsiveness that has potentially confounded studies of bone adaptation and translation of insights from our field. © 2018 The Authors. JBMR Plus Published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sundar Srinivasan
- Department of Orthopaedics and Sports Medicine University of Washington Seattle WA USA
| | - Danica Balsiger
- Department of Orthopaedics and Sports Medicine University of Washington Seattle WA USA
| | - Phillipe Huber
- Department of Orthopaedics and Sports Medicine University of Washington Seattle WA USA
| | - Brandon J Ausk
- Department of Orthopaedics and Sports Medicine University of Washington Seattle WA USA
| | - Steven D Bain
- Department of Orthopaedics and Sports Medicine University of Washington Seattle WA USA
| | - Edith M Gardiner
- Department of Orthopaedics and Sports Medicine University of Washington Seattle WA USA
| | - Ted S Gross
- Department of Orthopaedics and Sports Medicine University of Washington Seattle WA USA
| |
Collapse
|
47
|
Influence of Vitamin D Status and Mechanical Loading on the Morphometric and Mechanical Properties of the Mouse Tibia. J Med Biol Eng 2018. [DOI: 10.1007/s40846-018-0433-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
48
|
Javaheri B, Carriero A, Wood M, De Souza R, Lee PD, Shefelbine S, Pitsillides AA. Transient peak-strain matching partially recovers the age-impaired mechanoadaptive cortical bone response. Sci Rep 2018; 8:6636. [PMID: 29703931 PMCID: PMC5924380 DOI: 10.1038/s41598-018-25084-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022] Open
Abstract
Mechanoadaptation maintains bone mass and architecture; its failure underlies age-related decline in bone strength. It is unclear whether this is due to failure of osteocytes to sense strain, osteoblasts to form bone or insufficient mechanical stimulus. Mechanoadaptation can be restored to aged bone by surgical neurectomy, suggesting that changes in loading history can rescue mechanoadaptation. We use non-biased, whole-bone tibial analyses, along with characterisation of surface strains and ensuing mechanoadaptive responses in mice at a range of ages, to explore whether sufficient load magnitude can activate mechanoadaptation in aged bone. We find that younger mice adapt when imposed strains are lower than in mature and aged bone. Intriguingly, imposition of short-term, high magnitude loading effectively primes cortical but not trabecular bone of aged mice to respond. This response was regionally-matched to highest strains measured by digital image correlation and to osteocytic mechanoactivation. These data indicate that aged bone’s loading response can be partially recovered, non-invasively by transient, focal high strain regions. Our results indicate that old murine bone does respond to load when the loading is of sufficient magnitude, and bones’ age-related adaptation failure may be due to insufficient mechanical stimulus to trigger mechanoadaptation.
Collapse
Affiliation(s)
- Behzad Javaheri
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| | - Alessandra Carriero
- The City College of New York, Department of Biomedical Engineering, 160 Convent Avenue, New York, NY, 10031, USA
| | - Maria Wood
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Roberto De Souza
- Universidade Federal de Mato Grosso (UFMT), Departamento de Clínica, Av. Fernando Corrêa da Costa, 2367 - Boa Esperança, Cuiabá, 78060-900, Brazil
| | - Peter D Lee
- Manchester X-Ray Imaging Facility, University of Manchester, Research Complex at Harwell, RAL, Didcot, OX11 0FA, UK
| | - Sandra Shefelbine
- Department of Mechanical and Industrial Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Andrew A Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|
49
|
Liu C, Carrera R, Flamini V, Kenny L, Cabahug-Zuckerman P, George BM, Hunter D, Liu B, Singh G, Leucht P, Mann KA, Helms JA, Castillo AB. Effects of mechanical loading on cortical defect repair using a novel mechanobiological model of bone healing. Bone 2018; 108:145-155. [PMID: 29305998 PMCID: PMC8262576 DOI: 10.1016/j.bone.2017.12.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 12/20/2017] [Accepted: 12/29/2017] [Indexed: 12/30/2022]
Abstract
Mechanical loading is an important aspect of post-surgical fracture care. The timing of load application relative to the injury event may differentially regulate repair depending on the stage of healing. Here, we used a novel mechanobiological model of cortical defect repair that offers several advantages including its technical simplicity and spatially confined repair program, making effects of both physical and biological interventions more easily assessed. Using this model, we showed that daily loading (5N peak load, 2Hz, 60 cycles, 4 consecutive days) during hematoma consolidation and inflammation disrupted the injury site and activated cartilage formation on the periosteal surface adjacent to the defect. We also showed that daily loading during the matrix deposition phase enhanced both bone and cartilage formation at the defect site, while loading during the remodeling phase resulted in an enlarged woven bone regenerate. All loading regimens resulted in abundant cellular proliferation throughout the regenerate and fibrous tissue formation directly above the defect demonstrating that all phases of cortical defect healing are sensitive to physical stimulation. Stress was concentrated at the edges of the defect during exogenous loading, and finite element (FE)-modeled longitudinal strain (εzz) values along the anterior and posterior borders of the defect (~2200με) was an order of magnitude larger than strain values on the proximal and distal borders (~50-100με). It is concluded that loading during the early stages of repair may impede stabilization of the injury site important for early bone matrix deposition, whereas loading while matrix deposition and remodeling are ongoing may enhance stabilization through the formation of additional cartilage and bone.
Collapse
Affiliation(s)
- Chao Liu
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, USA; Department of Orthopaedic Surgery, New York University Langone Health, NYU Langone Orthopedic Hospital, NY, USA; Department of Veterans Affairs New York Harbor Healthcare System, New York, NY, USA
| | - Robert Carrera
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Vittoria Flamini
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, USA
| | - Lena Kenny
- Department of Orthopaedic Surgery, New York University Langone Health, NYU Langone Orthopedic Hospital, NY, USA
| | - Pamela Cabahug-Zuckerman
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, USA; Department of Orthopaedic Surgery, New York University Langone Health, NYU Langone Orthopedic Hospital, NY, USA; Department of Veterans Affairs New York Harbor Healthcare System, New York, NY, USA
| | - Benson M George
- Department of Surgery, Division of Plastic Surgery, Stanford University, Stanford, CA, USA
| | - Daniel Hunter
- Department of Surgery, Division of Plastic Surgery, Stanford University, Stanford, CA, USA
| | - Bo Liu
- Department of Surgery, Division of Plastic Surgery, Stanford University, Stanford, CA, USA
| | - Gurpreet Singh
- Department of Surgery, Division of Plastic Surgery, Stanford University, Stanford, CA, USA
| | - Philipp Leucht
- Department of Orthopaedic Surgery, New York University Langone Health, NYU Langone Orthopedic Hospital, NY, USA; Department of Cell Biology, New York University, New York, NY, USA
| | - Kenneth A Mann
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jill A Helms
- Department of Surgery, Division of Plastic Surgery, Stanford University, Stanford, CA, USA
| | - Alesha B Castillo
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, USA; Department of Orthopaedic Surgery, New York University Langone Health, NYU Langone Orthopedic Hospital, NY, USA; Department of Veterans Affairs New York Harbor Healthcare System, New York, NY, USA.
| |
Collapse
|
50
|
Sun D, Brodt MD, Zannit HM, Holguin N, Silva MJ. Evaluation of loading parameters for murine axial tibial loading: Stimulating cortical bone formation while reducing loading duration. J Orthop Res 2018; 36:682-691. [PMID: 28888055 PMCID: PMC5839947 DOI: 10.1002/jor.23727] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/31/2017] [Indexed: 02/04/2023]
Abstract
Classic studies in bone mechanobiology have established the importance of loading parameters on the anabolic response. Most of these early studies were done using loading methods not currently in favor, and using non-murine species. Our objective was to re-examine the effects of several loading parameters on the response of cortical bone using the contemporary murine axial tibial compression model. We subjected tibias of 5-month old, female C57Bl/6 mice to cyclic (4 Hz) mechanical loading and examined bone formation responses using dynamic and static histomorphometry. First, using a reference protocol of 1,200 cycles/day, 5 days/week for 2 weeks, we confirmed the significant influence of peak strain magnitude on periosteal mineralizing surface (Ps.MS/BS) and bone formation rate (Ps.BFR/BS) (p < 0.05, ANOVA). There was a significant induction of periosteal lamellar bone at a lower threshold of approx. -1,000 μϵ and a transition from lamellar-woven bone near -2,000 μϵ. In contrast, on the endocortical surface, bone formation indices did not exhibit a load magnitude-dependent response and no incidence of woven bone. Next, we found that reducing daily cycle number from 1,200 to 300 to 60 did not diminish the bone formation response (p > 0.05). On the other hand, reducing the daily frequency of loading from 5 consecutive days/week to 3 alternate days/week significantly diminished the periosteal response, from a loading-induced increase in Ps.MS/BS of 38% (loaded vs. control) for 5 days/week to only 15% for 3 days/week (p < 0.05). Finally, we determined that reducing the study duration from 2 to 1 weeks of loading did not affect bone formation outcomes. In conclusion, cyclic loading to -1,800 μϵ peak strain, at 4 Hz and 60 cycles/day for 5 consecutive days (1 week) induces an increase in periosteal lamellar bone formation with minimal incidence of woven bone in 5-month-old C57Bl/6 female mice. Our results provide a basis for reduction of loading duration (daily cycles and study length) without loss of anabolic effect as measured by dynamic histomorphometry. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:682-691, 2018.
Collapse
Affiliation(s)
- David Sun
- Department of Orthopaedic Surgery, Washington University in Saint
Louis, Saint Louis, Missouri,Department of Biomedical Engineering, Washington University in Saint
Louis, Saint Louis, Missouri
| | - Michael D. Brodt
- Department of Orthopaedic Surgery, Washington University in Saint
Louis, Saint Louis, Missouri
| | - Heather M. Zannit
- Department of Orthopaedic Surgery, Washington University in Saint
Louis, Saint Louis, Missouri,Department of Biomedical Engineering, Washington University in Saint
Louis, Saint Louis, Missouri
| | - Nilsson Holguin
- Department of Orthopaedic Surgery, Washington University in Saint
Louis, Saint Louis, Missouri
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University in Saint
Louis, Saint Louis, Missouri,Department of Biomedical Engineering, Washington University in Saint
Louis, Saint Louis, Missouri,Correspondence To: Matthew J. Silva, Ph.D.,
, Dept. of Orthopaedic Surgery/Campus Box
8233, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO
63110
| |
Collapse
|