1
|
Wang C, Fan M, Heo SC, Adams SM, Li T, Liu Y, Li Q, Loebel C, Burdick JA, Lu XL, Birk DE, Alisafaei F, Mauck RL, Han L. Structure, Mechanics, and Mechanobiology of Fibrocartilage Pericellular Matrix Mediated by Type V Collagen. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14750. [PMID: 40407177 DOI: 10.1002/advs.202414750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/01/2025] [Indexed: 05/28/2025]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding cells in various tissues, regulating matrix turnover, cell-matrix interactions, and disease. This study elucidates the structure-mechanical properties and mechanobiology of the PCM in fibrocartilage, using the murine meniscus as the model. The fibrocartilage PCM is comprised of thin, randomly oriented collagen fibrils that entrap proteoglycans, contrasting with the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). Compared to the ECM, the PCM exhibits lower modulus and greater isotropy, but has similar relative viscoelastic properties. In Col5a1+/- menisci, the reduction of collagen V results in thicker, more heterogeneous collagen fibrils, reduced modulus, loss of isotropy and faster viscoelastic relaxation in the PCM. Such altered PCM leads to impaired matrix-to-cell strain transmission, and in turn, disrupts mechanotransduction of meniscal cells, as illustrated by reduced calcium signaling activities and alters expression of matrix genes. In vitro, Col5a1+/- cells produce a weakened PCM with inferior properties and reduced protection of cells against tensile stretch. These findings highlight the PCM as a distinctive microstructure in fibrocartilage mechanobiology, underscoring a pivotal role of collagen V in PCM function. Targeting the PCM or its constituents offers potential for improving meniscus regeneration, osteoarthritis intervention and broader fibrocartilage-related therapies.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA, 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
2
|
Gauthier R, Follet H, Olivier C, Lemaire T, Mitton D, Peyrin F. Human cortical bone intrinsic permeability distribution based on 3D canalicular morphology. Bone 2025; 194:117441. [PMID: 40024426 DOI: 10.1016/j.bone.2025.117441] [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: 10/16/2024] [Revised: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Bone permeability is a key parameter that drives osteocyte-based mechanobiological modelling and remodelling. While previous experimental and numerical studies have estimated bone permeability based on the morphology of the lacuno-canalicular network, these studies often relied on simplified geometries. In the current study, bone permeability was characterized using more realistic canalicular geometry for the morphological data. Bone samples harvested from 27 human femoral bones were investigated using synchrotron radiation-based nano-computed tomography with a voxel size of 100 nm. After segmenting the canaliculi and lacunae, each canaliculus was investigated individually by applying a distance map and watershed algorithms. Bone permeability based on canalicular morphology was then assessed using the Kozeny relation, which defines the permeability of a porous medium with capillary-like pores. An averaged intrinsic permeability value of 8.8 10-18 m2 was obtained. It should be noted that this study considered an empty canalicular network, however in vivo, both cellular and peri-cellular matrices decrease space for interstitial fluid flow and thus permeability. Furthermore, a voxel size of 100 nm does not allow for the detection of smaller canaliculi, which may also modify average permeability. With the current data set and the analytic process applied, the results showed a heterogeneous permeability distribution within bone tissue, both when comparing osteonal and interstitial tissues and within an individual osteon. A difference was observed between male and female samples, and permeability appeared to significantly decrease with age. Finally, a significant correlation was found between permeability and canalicular length density, defined as canalicular length per unit bone volume. This study proposes a new form of the Kozeny law to express bone canalicular permeability as a proportional relationship with the canalicular length density. Importantly, this parameter can be directly quantified through confocal fluorescence microcopy, which is more convenient than synchrotron radiation-based nano-computed tomography. In conclusion, the current study confirms that confocal microscopy can be serve as a reliable tool to estimate bone permeability. However, the permeability values calculated here are solely based on canalicular morphology and do not consider cellular and peri-cellular intra-canalicular features.
Collapse
Affiliation(s)
- Remy Gauthier
- Univ Lyon, CNRS, INSA Lyon, Universite Claude Bernard Lyon 1, MATEIS UMR 5510, 69621 Villeurbanne, France.
| | - Hélène Follet
- Univ Lyon, Universite Claude Bernard Lyon 1, INSERM, LYOS UMR1033, Lyon, France
| | - Cécile Olivier
- Université Grenoble Alpes, INSERM UA7 Synchrotron Radiation for Biomedicine, Saint-Martin d'Hères, France
| | - Thibault Lemaire
- MSME UMR 8208, Univ Paris Est Créteil, Univ Gustave Eiffel, CNRS, 94010 Créteil, France
| | - David Mitton
- Univ Lyon, Univ Gustave Eiffel, Universite Claude Bernard Lyon 1, LBMC UMR_T9406, 69622 Lyon, France
| | - Francoise Peyrin
- Univ Lyon, INSA Lyon, Universite Claude Bernard Lyon 1, UJM-Saint Etienne, CNRS UMR 5220, Inserm U1206, CREATIS, 69621 Lyon, France
| |
Collapse
|
3
|
Yu W, Ou R, Hou Q, Li C, Yang X, Ma Y, Wu X, Chen W. Multiscale interstitial fluid computation modeling of cortical bone to characterize the hydromechanical stimulation of lacunar-canalicular network. Bone 2025; 193:117386. [PMID: 39746592 DOI: 10.1016/j.bone.2024.117386] [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: 10/10/2024] [Revised: 12/27/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Bone tissue is a biological composite material with a complex hierarchical structure that could continuously adjust its internal structure to adapt to the alterations in the external load environment. The fluid flow within bone is the main route of osteocyte metabolism, and the pore pressure as well as the fluid shear stress generated by it are important mechanical stimuli perceived by osteocytes. Owing to the irregular multiscale structure of bone tissue, the fluid stimulation that lacunar-canalicular network (LCN) in different regions of the tissue underwent remained unclear. In this study, we constructed a multiscale conduction model of fluid flow stimulus signals in bone tissue based on the poroelasticity theory. We analyzed the fluid flow behaviors at the macro-scale (whole bone tissue), macro-meso scale (periosteum, interstitial bone, osteon and endosteum), and micro-scale (lacunar-osteocyte-canalicular) levels. We explored how fluid stimulation at the tissue level correlated with that at the cellular level in cortical bone and characterized the distributions of the pore pressure, fluid velocity and fluid shear stress that the osteocytes experienced across the entire tissue structure. The results showed that the initial conditions of intramedullary pressure had a significant impact on the pore pressure of Haversian systems, but had a relatively small influence on the fluid velocity. The osteocyte which were located at different positions in the bone tissue received very distinct fluid stimuli. Osteocytes in the vicinity of the Haversian Canals experienced higher fluid shear stress stimulation. When the permeability of the LCN was within the range from 10-21 m2 to 10-18 m2, the distribution of pressure, fluid velocity and fluid shear stress within the osteon near the periosteum and endosteum was significantly different from that in other parts of the bone. However, when the permeability was less than 10-22 m2, such a difference did not exist. Particularly, the flow velocity at the lacunae was markedly higher than that in the canaliculi. Meanwhile, the pore pressure and fluid shear stress were conspicuously lower than those in the canaliculi. In this study, we considered the interconnections of different biofunctional units at different scales of bone tissue, construct a more complete multiscale model of bone tissue, and propose that osteocytes at different locations receive different fluid stimuli, which provides a reference for a deeper understanding of bone mechanotransduction.
Collapse
Affiliation(s)
- WeiLun Yu
- College of Biomedical Engineering, Jilin Medical University, Jilin, Jilin Province, China; Orthopedics, Shanxi Bethune Hospital, Taiyuan, Shanxi Province, China; College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - RenXia Ou
- College of Biomedical Engineering, Jilin Medical University, Jilin, Jilin Province, China
| | - Qi Hou
- College of Biomedical Engineering, Jilin Medical University, Jilin, Jilin Province, China
| | - ChunMing Li
- Orthopedics, Jilin Central Hospital, Jilin, Jilin Province, China
| | - XiaoHang Yang
- College of Biomedical Engineering, Jilin Medical University, Jilin, Jilin Province, China.
| | - YingHui Ma
- College of Biomedical Engineering, Jilin Medical University, Jilin, Jilin Province, China
| | - XiaoGang Wu
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi, China
| | - WeiYi Chen
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, Shanxi, China
| |
Collapse
|
4
|
Zhao W, Qian J, Li J, Su T, Deng X, Fu Y, Liang X, Cui H. From death to birth: how osteocyte death promotes osteoclast formation. Front Immunol 2025; 16:1551542. [PMID: 40165960 PMCID: PMC11955613 DOI: 10.3389/fimmu.2025.1551542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Bone remodeling is a dynamic and continuous process involving three components: bone formation mediated by osteoblasts, bone resorption mediated by osteoclasts, and bone formation-resorption balancing regulated by osteocytes. Excessive osteocyte death is found in various bone diseases, such as postmenopausal osteoporosis (PMOP), and osteoclasts are found increased and activated at osteocyte death sites. Currently, apart from apoptosis and necrosis as previously established, more forms of cell death are reported, including necroptosis, ferroptosis and pyroptosis. These forms of cell death play important role in the development of inflammatory diseases and bone diseases. Increasing studies have revealed that various forms of osteocyte death promote osteoclast formation via different mechanism, including actively secreting pro-inflammatory and pro-osteoclastogenic cytokines, such as tumor necrosis factor alpha (TNF-α) and receptor activator of nuclear factor-kappa B ligand (RANKL), or passively releasing pro-inflammatory damage associated molecule patterns (DAMPs), such as high mobility group box 1 (HMGB1). This review summarizes the established and potential mechanisms by which various forms of osteocyte death regulate osteoclast formation, aiming to provide better understanding of bone disease development and therapeutic target.
Collapse
Affiliation(s)
- Weijie Zhao
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Jiale Qian
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Ji Li
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Tian Su
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, College of pharmacy, Hainan Medical University, Haikou, China
| | - Xiaozhong Deng
- Department of Pain Treatment, Nanxi Shan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Yonghua Fu
- Department of Hand and Foot Microsurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xuelong Liang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongwang Cui
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Emergency Surgery, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
5
|
Guerra RM, Fowler VM, Wang L. Osteocyte Dendrites: How Do They Grow, Mature, and Degenerate in Mineralized Bone? Cytoskeleton (Hoboken) 2024. [PMID: 39651620 DOI: 10.1002/cm.21964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024]
Abstract
Osteocytes, the most abundant bone cells, form an extensive cellular network via interconnecting dendrites. Like neurons in the brain, the long-lived osteocytes perceive mechanical and biological inputs and signal to other effector cells, leading to the homeostasis and turnover of bone tissues. Despite the appreciation of osteocytes' vital roles in bone biology, the initiation, growth, maintenance, and eventual degradation of osteocyte dendrites are poorly understood due to their full encasement by mineralized matrix. With the advancement of imaging modalities and genetic models, the architectural organization and molecular composition of the osteocyte dendrites, as well as their morphological changes with aging and diseases, have begun to be revealed. However, several long-standing mysteries remain unsolved, including (1) how the dendrites are initiated and elongated when a surface osteoblast becomes embedded as an osteocyte; (2) how the dendrites maintain a relatively stable morphology during their decades-long life span; (3) what biological processes control the dendrite morphology, connectivity, and stability; and (4) if these processes are influenced by age, sex, hormones, and mechanical loading. Our review of long, thin actin filament (F-actin)-containing processes extending from other cells leads to a working model that serves as a starting point to investigate the formation and maintenance of osteocyte dendrites and their degradation with aging and diseases.
Collapse
Affiliation(s)
- Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Velia M Fowler
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Liyun Wang
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
6
|
Knani L, Venditti M, Rouis H, Minucci S, Messaoudi I. Effects of dopaminergic neuron degeneration on osteocyte apoptosis and osteogenic markers in 6-OHDA male rat model of Parkinson's disease. Bone 2024; 190:117271. [PMID: 39369834 DOI: 10.1016/j.bone.2024.117271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/22/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Parkinson's disease (PD) and osteoporosis are prevalent chronic conditions that impact a significant proportion of the aging population. Observational and longitudinal studies consistently demonstrate that individuals with PD face an elevated risk of osteoporosis and reduced bone mineral density compared to control groups. However, there is currently no experimental evidence demonstrating the impact of dopaminergic neuron degeneration on bone metabolism. In the present study, we used a male rat model of PD induced by unilateral injection of 6-hydroxydopamine (6-OHDA) in the left medial forebrain bundle (MFB) to evaluate the effect of dopaminergic neuron lesion on certain parameters of bone metabolism. To confirm the dopaminergic neuron lesion, cylinder and Rotarod tests were applied to rats injected with 6-OHDA or vehicle. Osteocyte density and viability were determined through histology and TUNEL assay. Western Blot and immunohistochemistry analysis were performed to investigate whether dopaminergic degeneration influences the expression of some apoptotic markers (Caspase 3 and Cytochrome C) and some osteogenic markers (ALP, OCN, and RUNX2). Our findings show that the dopaminergic lesion resulting from the injection of 6-OHDA was successfully confirmed through behavioral tests. Furthermore, the degeneration of dopaminergic neurons induced by 6-OHDA leads to apoptosis of osteocytes associated with a significant reduction in the tissue expression of the studied osteogenic markers. Thus, our study provides evidence that 6-OHDA-induced degeneration of dopaminergic neurons leads to osteocyte apoptosis, which may contribute to the development of some signs of osteoporosis.
Collapse
Affiliation(s)
- Latifa Knani
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut Supérieur de Biotechnologie, Université de Monastir, Monastir, Tunisia.
| | - Massimo Venditti
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate "F. Bottazzi", Università della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Hajer Rouis
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut Supérieur de Biotechnologie, Université de Monastir, Monastir, Tunisia
| | - Sergio Minucci
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate "F. Bottazzi", Università della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Imed Messaoudi
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut Supérieur de Biotechnologie, Université de Monastir, Monastir, Tunisia
| |
Collapse
|
7
|
Qin L, Yang S, Zhao C, Yang J, Li F, Xu Z, Yang Y, Zhou H, Li K, Xiong C, Huang W, Hu N, Hu X. Prospects and challenges for the application of tissue engineering technologies in the treatment of bone infections. Bone Res 2024; 12:28. [PMID: 38744863 PMCID: PMC11094017 DOI: 10.1038/s41413-024-00332-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/08/2024] [Accepted: 04/01/2024] [Indexed: 05/16/2024] Open
Abstract
Osteomyelitis is a devastating disease caused by microbial infection in deep bone tissue. Its high recurrence rate and impaired restoration of bone deficiencies are major challenges in treatment. Microbes have evolved numerous mechanisms to effectively evade host intrinsic and adaptive immune attacks to persistently localize in the host, such as drug-resistant bacteria, biofilms, persister cells, intracellular bacteria, and small colony variants (SCVs). Moreover, microbial-mediated dysregulation of the bone immune microenvironment impedes the bone regeneration process, leading to impaired bone defect repair. Despite advances in surgical strategies and drug applications for the treatment of bone infections within the last decade, challenges remain in clinical management. The development and application of tissue engineering materials have provided new strategies for the treatment of bone infections, but a comprehensive review of their research progress is lacking. This review discusses the critical pathogenic mechanisms of microbes in the skeletal system and their immunomodulatory effects on bone regeneration, and highlights the prospects and challenges for the application of tissue engineering technologies in the treatment of bone infections. It will inform the development and translation of antimicrobial and bone repair tissue engineering materials for the management of bone infections.
Collapse
Affiliation(s)
- Leilei Qin
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Shuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Chen Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Jianye Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Feilong Li
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Zhenghao Xu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Yaji Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Kainan Li
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan, 610081, China
| | - Chengdong Xiong
- University of Chinese Academy of Sciences, Bei Jing, 101408, China
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan, 610081, China.
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
8
|
Niroobakhsh M, Laughrey LE, Dallas SL, Johnson ML, Ganesh T. Computational modeling based on confocal imaging predicts changes in osteocyte and dendrite shear stress due to canalicular loss with aging. Biomech Model Mechanobiol 2024; 23:129-143. [PMID: 37642807 DOI: 10.1007/s10237-023-01763-w] [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: 01/30/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
Exercise and physical activity exert mechanical loading on the bones which induces bone formation. However, the relationship between the osteocyte lacunar-canalicular morphology and mechanical stress experienced locally by osteocytes transducing signals for bone formation is not fully understood. In this study, we used computational modeling to predict the effect of canalicular density, the number of fluid inlets, and load direction on fluid flow shear stress (FFSS) and bone strains and how these might change following the microstructural deterioration of the lacunar-canalicular network that occurs with aging. Four distinct computational models were initially generated of osteocytes with either ten or eighteen dendrites using a fluid-structure interaction method with idealized geometries. Next, a young and a simulated aged osteocyte were developed from confocal images after FITC staining of the femur of a 4-month-old C57BL/6 mouse to estimate FFSS using a computational fluid dynamics approach. The models predicted higher fluid velocities in the canaliculi versus the lacunae. Comparison of idealized models with five versus one fluid inlet indicated that with four more inlets, one-half of the dendrites experienced FFSS greater than 0.8 Pa, which has been associated with osteogenic responses. Confocal image-based models of real osteocytes indicated a six times higher ratio of canalicular to lacunar surface area in the young osteocyte model than the simulated aged model and the average FFSS in the young model (FFSS = 0.46 Pa) was three times greater than the aged model (FFSS = 0.15 Pa). Interestingly, the surface area with FFSS values above 0.8 Pa was 23 times greater in the young versus the simulated aged model. These findings may explain the impaired mechano-responsiveness of osteocytes with aging.
Collapse
Affiliation(s)
- Mohammad Niroobakhsh
- Division of Natural and Built Environment, School of Science and Engineering, University of Missouri-Kansas City, 350 L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO, 64110, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Loretta E Laughrey
- Division of Natural and Built Environment, School of Science and Engineering, University of Missouri-Kansas City, 350 L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO, 64110, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Thiagarajan Ganesh
- Division of Natural and Built Environment, School of Science and Engineering, University of Missouri-Kansas City, 350 L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO, 64110, USA.
| |
Collapse
|
9
|
Wu W, Brandt C, Zhou X, Tang S. Label-free multimodal imaging with simultaneous two-photon and three-photon microscopy and kernel-based nonlinear scaling denoising. BIOMEDICAL OPTICS EXPRESS 2024; 15:114-130. [PMID: 38223188 PMCID: PMC10783916 DOI: 10.1364/boe.504550] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024]
Abstract
We report on a compact multimodal imaging system that can acquire two-photon microscopy (2PM) and three-photon microscopy (3PM) images simultaneously. With dual excitation wavelengths, multiple contrasts including two-photon-excitation-fluorescence (2PEF), second harmonic generation (SHG), and third harmonic generation (THG) are acquired simultaneously from cells, collagen fibers, and interfaces, all label-free. Challenges related to the excitation by two wavelengths and the effective separation of 2PM and 3PM signals are discussed and addressed. The data processing challenge where multiple contrasts can have significantly varying signal levels is also addressed. A kernel-based nonlinear scaling (KNS) denoising method is introduced to reduce noise from ultra-low signal images and generate high-quality multimodal images. Simultaneous 2PM and 3PM imaging is demonstrated on various tissue samples. The simultaneous acquisition speeds up the imaging process and minimizes the commonly encountered problem of motion artifacts and mechanical drift in sequential acquisition. Multimodal imaging with simultaneous 2PM and 3PM will have great potential for label-free in-vivo imaging of biological tissues.
Collapse
Affiliation(s)
- Wentao Wu
- Department of Electrical and Computer Engineering, University of British Columbia, 5500-2332 Main Mall, Vancouver, BC V6 T 1Z4, Canada
| | - Christoph Brandt
- Department of Electrical and Computer Engineering, University of British Columbia, 5500-2332 Main Mall, Vancouver, BC V6 T 1Z4, Canada
| | - Xin Zhou
- Department of Electrical and Computer Engineering, University of British Columbia, 5500-2332 Main Mall, Vancouver, BC V6 T 1Z4, Canada
| | - Shuo Tang
- Department of Electrical and Computer Engineering, University of British Columbia, 5500-2332 Main Mall, Vancouver, BC V6 T 1Z4, Canada
| |
Collapse
|
10
|
Zhao F, Zhang Y, Pei S, Wang S, Hu L, Wang L, Qian A, Yang TL, Guo Y. Mechanobiological crosstalk among bone cells and between bone and other organs. BONE CELL BIOMECHANICS, MECHANOBIOLOGY AND BONE DISEASES 2024:215-247. [DOI: 10.1016/b978-0-323-96123-3.00015-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
11
|
Bahadorani F, Hadadzadeh H, Mirahmadi-Zare SZ, Masaeli E. Nanocore-Shell Bone Filler Contained Mesoporous Silica Modified with Hydroxyapatite Precursors; Wrapped in a Natural Metal-Phenolic Network. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:16090-16100. [PMID: 37921536 DOI: 10.1021/acs.langmuir.3c02227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Various therapeutic strategies have been developed to address bone diseases caused by aging society and skeletal defects caused by trauma or accidental events. One such approach is using bone fillers, such as hydroxyapatite (HA) and bioactive glasses. Although they have provided effective osteogenesis, infection and inflammation due to the surgical procedure and uncontrolled ion release can hinder the efficiency of bone regeneration. In response to these challenges, immobilizing a neutral metal-phenolic network on the surface of osteoconductive nanoparticles would be the master key to achieving a gradual, controlled release during the mineralization period and reducing infection and inflammation through biological pathways. In this regard, a mesoporous silica nanocomposite modified by an HA precursor was synthesized to enhance bone regeneration. In addition, to improve the therapeutic effects, its surface was wrapped with a magnesium-phenolic network made from pomegranate extract, which can simultaneously produce anti-inflammatory and antibacterial effects. The obtained core-shell nanocomposite was characterized by its physicochemical properties, biocompatibility, and bioactivity. The in vitro studies revealed that the synthesized nanocomposite exhibits higher osteogenic activity than the control groups, as confirmed by alizarin red staining. Moreover, the nanocomposite maintained low toxicity as measured by the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay and increased antibacterial activity against Staphylococcus aureus and Escherichia coli compared with the control groups. Therefore, this research presents a promising strategy for bone regeneration, combining the advantages of mesoporous silica nanocomposite modified by an HA precursor with the beneficial effects of a magnesium-phenolic network.
Collapse
Affiliation(s)
- Fatemeh Bahadorani
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Hassan Hadadzadeh
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Seyede Zohreh Mirahmadi-Zare
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | - Elahe Masaeli
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| |
Collapse
|
12
|
Ahmed F, Minamizaki T, Aubin JE, Damayanti MA, Yoshiko Y. Large scale analysis of osteocyte lacunae in klotho hypomorphic mice using high-resolution micro-computed tomography. Ann Anat 2023; 250:152142. [PMID: 37572763 DOI: 10.1016/j.aanat.2023.152142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Osteocytes are the most abundant cell type in adult bone, and the morphological characteristics of osteocytes and their lacunae appear to influence bone mass and fragility. Although conventional computed tomography (CT) has contributed greatly to advances in bone morphometry, capturing details of the entire hierarchical assembly, e.g., osteocyte lacuna parameters, has been limited by the analytical performance of CT (> 1 µm resolution). METHODS We used high-resolution (700 nm) micro-CT to evaluate and compare the osteocyte lacuna parameters over a large scale, i.e., in a maximum of about 45,700 lacunae (average), in tibial metaphyseal cortical bones of wild-type (WT) and αKlotho-hypomorphic (kl/kl) mice, the latter a model that exhibits osteopenia and aberrant osteocytes. RESULTS Of osteocyte lacuna parameters, lacunar surface per lacunar volume were significantly lower and lacuna diameter were significantly larger in kl/kl mice compared to WT mice. By analysis of individual osteocyte lacunae, we found that lacunar sphericity in kl/kl mice was higher than that in WT mice, and the diameters in the major and the minor axes were respectively lower and higher in kl/kl mice, especially at the proximal site of the region of interest. CONCLUSION We successfully assessed osteocyte lacuna parameters on the largest scale in mice reported to date and found that the shape of osteocyte lacunae of kl/kl mice are significantly different from those of WT mice. Although the mechanisms underlying the lacunar shape differences observed are not yet clear, changes in lacunar geometry are known to affect the transitions of strains to the osteocyte microenvironment and likely local osteocyte response(s). Thus, the fact that the differences are limited to the mesial region near the primary spongiosa suggests the likelihood of site-specific anomalies in mechanosensitive effects in kl/kl osteocytes with consequent site-specific effects bone metabolism and function.
Collapse
Affiliation(s)
- Faisal Ahmed
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Tomoko Minamizaki
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Jane E Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Merry Annisa Damayanti
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan; Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, Indonesia
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.
| |
Collapse
|
13
|
Liu Z, Wang Q, Zhang J, Qi S, Duan Y, Li C. The Mechanotransduction Signaling Pathways in the Regulation of Osteogenesis. Int J Mol Sci 2023; 24:14326. [PMID: 37762629 PMCID: PMC10532275 DOI: 10.3390/ijms241814326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Bones are constantly exposed to mechanical forces from both muscles and Earth's gravity to maintain bone homeostasis by stimulating bone formation. Mechanotransduction transforms external mechanical signals such as force, fluid flow shear, and gravity into intracellular responses to achieve force adaptation. However, the underlying molecular mechanisms on the conversion from mechanical signals into bone formation has not been completely defined yet. In the present review, we provide a comprehensive and systematic description of the mechanotransduction signaling pathways induced by mechanical stimuli during osteogenesis and address the different layers of interconnections between different signaling pathways. Further exploration of mechanotransduction would benefit patients with osteoporosis, including the aging population and postmenopausal women.
Collapse
Affiliation(s)
- Zhaoshuo Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Sihan Qi
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yingying Duan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
14
|
Lee BS, Murray C, Liu J, Kim M, Hwang MS, Yueh T, Mansour M, Qamar S, Agarwal G, Kim DG. The myosin and RhoGAP MYO9B influences osteocyte dendrite growth and responses to mechanical stimuli. Front Bioeng Biotechnol 2023; 11:1243303. [PMID: 37675403 PMCID: PMC10477788 DOI: 10.3389/fbioe.2023.1243303] [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: 06/20/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction: Myosin IXB (MYO9B) is an unconventional myosin with RhoGAP activity and thus is a regulator of actin cytoskeletal organization. MYO9B was previously shown to be necessary for skeletal growth and health and to play a role in actin-based functions of both osteoblasts and osteoclasts. However, its role in responses to mechanical stimulation of bone cells has not yet been described. Therefore, experiments were undertaken to determine the role of MYO9B in bone cell responses to mechanical stress both in vitro and in vivo. Methods: MYO9B expression was knocked down in osteoblast and osteocyte cell lines using RNA interference and the resulting cells were subjected to mechanical stresses including cyclic tensile strain, fluid shear stress, and plating on different substrates (no substrate vs. monomeric or polymerized collagen type I). Osteocytic cells were also subjected to MYO9B regulation through Slit-Robo signaling. Further, wild-type or Myo9b -/- mice were subjected to a regimen of whole-body vibration (WBV) and changes in bone quality were assessed by micro-CT. Results: Unlike control cells, MYO9B-deficient osteoblastic cells subjected to uniaxial cyclic tensile strain were unable to orient their actin stress fibers perpendicular to the strain. Osteocytic cells in which MYO9B was knocked down exhibited elongated dendrites but were unable to respond normally to treatments that increase dendrite length such as fluid shear stress and Slit-Robo signaling. Osteocytic responses to mechanical stimuli were also found to be dependent on the polymerization state of collagen type I substrates. Wild-type mice responded to WBV with increased bone tissue mineral density values while Myo9b -/- mice responded with bone loss. Discussion: These results demonstrate that MYO9B plays a key role in mechanical stress-induced responses of bone cells in vitro and in vivo.
Collapse
Affiliation(s)
- Beth S. Lee
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Cynthia Murray
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Jie Liu
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Minji Kim
- Department of Orthodontics, Graduate School of Clinical Dentistry, Ewha Womans University, Seoul, Republic of Korea
| | - Min Sik Hwang
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Tina Yueh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Myrna Mansour
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Sana Qamar
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Gunjan Agarwal
- Department of Mechanical and Aerospace Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
15
|
Detailing the influence of PEO-coated biodegradable Mg-based implants on the lacuno-canalicular network in sheep bone: A pilot study. Bioact Mater 2023; 26:14-23. [PMID: 36875051 PMCID: PMC9975618 DOI: 10.1016/j.bioactmat.2023.02.018] [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: 11/15/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
An increasing prevalence of bone-related injuries and aging geriatric populations continue to drive the orthopaedic implant market. A hierarchical analysis of bone remodelling after material implantation is necessary to better understand the relationship between implant and bone. Osteocytes, which are housed and communicate through the lacuno-canalicular network (LCN), are integral to bone health and remodelling processes. Therefore, it is essential to examine the framework of the LCN in response to implant materials or surface treatments. Biodegradable materials offer an alternative solution to permanent implants, which may require revision or removal surgeries. Magnesium alloys have resurfaced as promising materials due to their bone-like properties and safe degradation in vivo. To further tailor their degradation capabilities, surface treatments such as plasma electrolytic oxidation (PEO) have demonstrated to slow degradation. For the first time, the influence of a biodegradable material on the LCN is investigated by means of non-destructive 3D imaging. In this pilot study, we hypothesize noticeable variations in the LCN caused by altered chemical stimuli introduced by the PEO-coating. Utilising synchrotron-based transmission X-ray microscopy, we have characterised morphological LCN differences around uncoated and PEO-coated WE43 screws implanted into sheep bone. Bone specimens were explanted after 4, 8, and 12 weeks and regions near the implant surface were prepared for imaging. Findings from this investigation indicate that the slower degradation of PEO-coated WE43 induces healthier lacunar shapes within the LCN. However, the stimuli perceived by the uncoated material with higher degradation rates induces a greater connected LCN better prepared for bone disturbance.
Collapse
|
16
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
17
|
Tuncer C, Güden M, Orhan M, Sarıkaya MK, Taşdemirci A. Quasi-static and dynamic Brazilian testing and failure analysis of a deer antler in the transverse to the osteon growth direction. J Mech Behav Biomed Mater 2023; 138:105648. [PMID: 36610280 DOI: 10.1016/j.jmbbm.2023.105648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
The transverse tensile strength of a naturally fallen red deer antler (Cervus Elaphus) was determined through indirect Brazilian tests using dry disc-shape specimens at quasi-static and high strain rates. Dynamic Brazilian tests were performed in a compression Split-Hopkinson Pressure Bar. Quasi-static tensile and indirect Brazilian tests were also performed along the osteon growth direction for comparison. The quasi-static transverse tensile strength ranged 31.5-44.5 MPa. The strength increased to 83 MPa on the average in the dynamic Brazilian tests, proving a rate sensitive transverse strength. The quasi-static tensile strength in the osteon growth direction was however found comparably higher, 192 MPa. A Weibull analysis indicated a higher tensile ductility in the osteon growth direction than in the transverse to the osteon growth direction. The microscopic analysis of the quasi-static Brazilian test specimens (tensile strain along the osteon growth direction) revealed a micro-cracking mechanism operating by the crack deflection/twisting at the lacunae in the concentric lamellae region and at the interface between concentric lamellae and interstitial lamellae. On the other side, the specimens in the transverse direction fractured in a more brittle manner by the separation/delamination of the concentric lamellae and pulling of the interstitial lamellae. The detected increase in the transverse strength in the high strain rate tests was further ascribed to the pull and fracture of the visco-plastic collagen fibers in the interstitial lamellae. This was also confirmed microscopically; the dynamically tested specimens exhibited flatter fracture surfaces.
Collapse
Affiliation(s)
- Can Tuncer
- Department of Mechanical Engineering, Faculty of Engineering, Pamukkale University, Denizli, Turkey
| | - Mustafa Güden
- Department of Mechanical Engineering, Faculty of Engineering, İzmir Institute of Technology, Urla, İzmir, Turkey
| | - Mehmet Orhan
- Department of Mechanical Engineering, Faculty of Engineering, Pamukkale University, Denizli, Turkey.
| | - Mustafa Kemal Sarıkaya
- Department of Mechanical Engineering, Faculty of Engineering, İzmir Institute of Technology, Urla, İzmir, Turkey
| | - Alper Taşdemirci
- Department of Mechanical Engineering, Faculty of Engineering, İzmir Institute of Technology, Urla, İzmir, Turkey
| |
Collapse
|
18
|
Sato M, Shah FA. Contributions of Resin Cast Etching to Visualising the Osteocyte Lacuno-Canalicular Network Architecture in Bone Biology and Tissue Engineering. Calcif Tissue Int 2023; 112:525-542. [PMID: 36611094 PMCID: PMC10106349 DOI: 10.1007/s00223-022-01058-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/21/2022] [Indexed: 01/09/2023]
Abstract
Recent years have witnessed an evolution of imaging technologies towards sophisticated approaches for visualising cells within their natural environment(s) and for investigating their interactions with other cells, with adjacent anatomical structures, and with implanted biomaterials. Resin cast etching (RCE) is an uncomplicated technique involving sequential acid etching and alkali digestion of resin embedded bone to observe the osteocyte lacuno-canalicular network using scanning electron microscopy. This review summarises the applicability of RCE to bone and the bone-implant interface. Quantitative parameters such as osteocyte size, osteocyte density, and number of canaliculi per osteocyte, and qualitative metrics including osteocyte shape, disturbances in the arrangement of osteocytes and canaliculi, and physical communication between osteocytes and implant surfaces can be investigated. Ageing, osteoporosis, long-term immobilisation, spinal cord injury, osteoarthritis, irradiation, and chronic kidney disease have been shown to impact osteocyte lacuno-canalicular network morphology. In addition to titanium, calcium phosphates, and bioactive glass, observation of direct connectivity between osteocytes and cobalt chromium provides new insights into the osseointegration potential of materials conventionally viewed as non-osseointegrating. Other applications include in vivo and in vitro testing of polymer-based tissue engineering scaffolds and tissue-engineered ossicles, validation of ectopic osteochondral defect models, ex vivo organ culture of whole bones, and observing the effects of gene dysfunction/deletion on the osteocyte lacuno-canalicular network. Without additional contrast staining, any resin embedded specimen (including clinical biopsies) can be used for RCE. The multitude of applications described here attest to the versatility of RCE for routine use within correlative analytical workflows, particularly in biomaterials science.
Collapse
Affiliation(s)
- Mari Sato
- Oral Biochemistry and Molecular Biology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Furqan A Shah
- Department of Biomaterials, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
19
|
Wang JS, Wein MN. Pathways Controlling Formation and Maintenance of the Osteocyte Dendrite Network. Curr Osteoporos Rep 2022; 20:493-504. [PMID: 36087214 PMCID: PMC9718876 DOI: 10.1007/s11914-022-00753-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the molecular mechanisms involved in osteocyte dendrite formation, summarize the similarities between osteocytic and neuronal projections, and highlight the importance of osteocyte dendrite maintenance in human skeletal disease. RECENT FINDINGS It is suggested that there is a causal relationship between the loss of osteocyte dendrites and the increased osteocyte apoptosis during conditions including aging, microdamage, and skeletal disease. A few mechanisms are proposed to control dendrite formation and outgrowth, such as via the regulation of actin polymerization dynamics. This review addresses the impact of osteocyte dendrites in bone health and disease. Recent advances in multi-omics, in vivo and in vitro models, and microscopy-based imaging have provided novel approaches to reveal the underlying mechanisms that regulate dendrite development. Future therapeutic approaches are needed to target the process of osteocyte dendrite formation.
Collapse
Affiliation(s)
- Jialiang S Wang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
20
|
Buettmann EG, Goldscheitter GM, Hoppock GA, Friedman MA, Suva LJ, Donahue HJ. Similarities Between Disuse and Age-Induced Bone Loss. J Bone Miner Res 2022; 37:1417-1434. [PMID: 35773785 PMCID: PMC9378610 DOI: 10.1002/jbmr.4643] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/07/2022]
Abstract
Disuse and aging are known risk factors associated with low bone mass and quality deterioration, resulting in increased fracture risk. Indeed, current and emerging evidence implicate a large number of shared skeletal manifestations between disuse and aging scenarios. This review provides a detailed overview of current preclinical models of musculoskeletal disuse and the clinical scenarios they seek to recapitulate. We also explore and summarize the major similarities between bone loss after extreme disuse and advanced aging at multiple length scales, including at the organ/tissue, cellular, and molecular level. Specifically, shared structural and material alterations of bone loss are presented between disuse and aging, including preferential loss of bone at cancellous sites, cortical thinning, and loss of bone strength due to enhanced fragility. At the cellular level bone loss is accompanied, during disuse and aging, by increased bone resorption, decreased formation, and enhanced adipogenesis due to altered gap junction intercellular communication, WNT/β-catenin and RANKL/OPG signaling. Major differences between extreme short-term disuse and aging are discussed, including anatomical specificity, differences in bone turnover rates, periosteal modeling, and the influence of subject sex and genetic variability. The examination also identifies potential shared mechanisms underlying bone loss in aging and disuse that warrant further study such as collagen cross-linking, advanced glycation end products/receptor for advanced glycation end products (AGE-RAGE) signaling, reactive oxygen species (ROS) and nuclear factor κB (NF-κB) signaling, cellular senescence, and altered lacunar-canalicular connectivity (mechanosensation). Understanding the shared structural alterations, changes in bone cell function, and molecular mechanisms common to both extreme disuse and aging are paramount to discovering therapies to combat both age-related and disuse-induced osteoporosis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Galen M Goldscheitter
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Gabriel A Hoppock
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael A Friedman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Henry J Donahue
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
21
|
Cen H, Gong H, Liu H, Jia S, Wu X, Fan Y. A Comparative Study on the Multiscale Mechanical Responses of Human Femoral Neck Between the Young and the Elderly Using Finite Element Method. Front Bioeng Biotechnol 2022; 10:893337. [PMID: 35600894 PMCID: PMC9117745 DOI: 10.3389/fbioe.2022.893337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Femoral neck fracture (FNF) is the most serious bone disease in the elderly population. The multiscale mechanical response is a key to predicting the strength of the femoral neck, assessing the risk of FNF, and exploring the role of mechanosensation and mechanotransmission in bone remodeling, especially in the context of aging bone.Methods: Multiscale finite element (FE) models of the proximal femur for both young and elderly people were developed. The models included organ scale (proximal femur), tissue scale (cortical bone), tissue element scale (osteon), and cell scale [osteocyte lacuna-canalicular network (LCN) and extracellular matrix (ECM), OLCEM]. The mechanical responses of cortical bone and osteocytes in the mid-femoral neck and the differences in mechanical responses between these two scales were investigated.Results: The mechanical responses of cortical bone and osteocyte showed significant differences between the elderly and the young. The minimum principal strains and mean SEDs of cortical bone in the elderly were 2.067–4.708 times and 3.093–14.385 times of the values in the young, respectively; the minimum principal strains and mean SEDs of osteocyte in the elderly were 1.497–3.246 times and 3.044–12 times of the values in the young, respectively; the amplification factors of minimum principal strain in the inferior (Inf), anterior (Ant), and posterior (Post) quadrants in the young were 1.241–1.804 times of the values in the elderly, but the amplification factor of minimum principal strain in the superior (Sup) quadrant was 87.4% of the value in the elderly; the amplification factors of mean SED in the young were 1.124–9.637 times of the values in the elderly.Conclusion: The mass and bone mineral density (BMD) of cortical bone in the femoral neck is closely related to the mechanical response of osteocytes, which provides a new idea for improving cortical bone quality. Perhaps cortical bone quality could be improved by stimulating osteocytes. Quadrantal differences of bone quality in the mid-femoral neck should be considered to improve fracture risk prediction in the future.
Collapse
Affiliation(s)
- Haipeng Cen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - He Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: He Gong,
| | - Haibo Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shaowei Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xiaodan Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- School of Engineering Medicine, Beihang University, Beijing, China
| |
Collapse
|
22
|
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: 3.0] [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
|
23
|
Murshid SA. Bone permeability and mechanotransduction: Some current insights into the function of the lacunar-canalicular network. Tissue Cell 2022; 75:101730. [PMID: 35032785 DOI: 10.1016/j.tice.2022.101730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Abstract
Lacunar-canalicular (LC) permeability involves the passage of fluids, nutrients, oxygen, ions, and signalling molecules through bone tissue, facilitating the maintenance of bone vitality and function and responses to various physiological conditions and diseases. LC permeability and fluid flow-shear stress/drag force play important roles in mechanotransduction in bone tissue by inducing mechanical stimuli in osteocytes, modulating cellular functions, and determining bone adaptation. Alterations in LC structure may therefore influence the fluid flow pattern through the LC network, thereby affecting the ability of osteocytes to sense and translate mechanical signals and possibly contributing to bone remodelling. Several bone-health conditions are associated with changes in LC structure and function and may affect mechanotransduction and responses, although the mechanisms underlying these associations are still not fully understood. In this review, recent studies of LC networks, their formation and transfer mechanical stimuli, and changes in structure, functional permeability, and mechanotransduction that result from age, pathology, and mechanical loading are discussed. Additionally, applications of vibration and low-intensity pulsed ultrasound in bone healthcare and regeneration fields are also presented.
Collapse
Affiliation(s)
- Sakhr Ahmed Murshid
- Institute for Globally Distributed Open Research and Education (IGDORE); Ilmajoki Health Public Dental Clinics, Social and Health Care Services in Jalasjärvi, Ilmajoki, Kurikka, Finland.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Osteocytes are the conductors of bone adaptation and remodelling. Buried inside the calcified matrix, they sense mechanical cues and signal osteoclasts in case of low activity, and osteoblasts when stresses are high. How do osteocytes detect mechanical stress? What physical signal do they perceive? Finite element analysis is a useful tool to address these questions as it allows calculating stresses, strains and fluid flow where they cannot be measured. The purpose of this review is to evaluate the capabilities and challenges of finite element models of bone, in particular the osteocytes and load-induced activation mechanisms. RECENT FINDINGS High-resolution imaging and increased computational power allow ever more detailed modelling of osteocytes, either in isolation or embedded within the mineralised matrix. Over the years, homogeneous models of bone and osteocytes got replaced by heterogeneous and microstructural models, including, e.g. the lacuno-canalicular network and the cytoskeleton. The lacuno-canalicular network induces strain amplifications and the osteocyte protrusions seem to be stimulated much more than the cell body, both by strain and fluid flow. More realistic cell geometries, like minute constrictions of the canaliculi, increase this effect. Microstructural osteocyte models describe the transduction of external stimuli to the nucleus. Supracellular multiscale models (e.g. of a tunnelling osteon) allow to study differential loading of osteocytes and to distinguish between strain and fluid flow as the pivotal stimulatory cue. In the future, the finite element models may be enhanced by including chemical transport and intercellular communication between osteocytes, osteoclasts and osteoblasts.
Collapse
Affiliation(s)
- Theodoor H Smit
- Department of Medical Biology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Orthopaedic Surgery, Amsterdam Movement Sciences Research Institute, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Choi JUA, Kijas AW, Lauko J, Rowan AE. The Mechanosensory Role of Osteocytes and Implications for Bone Health and Disease States. Front Cell Dev Biol 2022; 9:770143. [PMID: 35265628 PMCID: PMC8900535 DOI: 10.3389/fcell.2021.770143] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Bone homeostasis is a dynamic equilibrium between bone-forming osteoblasts and bone-resorbing osteoclasts. This process is primarily controlled by the most abundant and mechanosensitive bone cells, osteocytes, that reside individually, within chambers of porous hydroxyapatite bone matrix. Recent studies have unveiled additional functional roles for osteocytes in directly contributing to local matrix regulation as well as systemic roles through endocrine functions by communicating with distant organs such as the kidney. Osteocyte function is governed largely by both biochemical signaling and the mechanical stimuli exerted on bone. Mechanical stimulation is required to maintain bone health whilst aging and reduced level of loading are known to result in bone loss. To date, both in vivo and in vitro approaches have been established to answer important questions such as the effect of mechanical stimuli, the mechanosensors involved, and the mechanosensitive signaling pathways in osteocytes. However, our understanding of osteocyte mechanotransduction has been limited due to the technical challenges of working with these cells since they are individually embedded within the hard hydroxyapatite bone matrix. This review highlights the current knowledge of the osteocyte functional role in maintaining bone health and the key regulatory pathways of these mechanosensitive cells. Finally, we elaborate on the current therapeutic opportunities offered by existing treatments and the potential for targeting osteocyte-directed signaling.
Collapse
Affiliation(s)
- Jung Un Ally Choi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Amanda W Kijas
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Jan Lauko
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Alan E Rowan
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Liu HY, Zhao CH, Zhang H, Wang W, Liu QJ. Simulation study on the effect of resistance exercise on the hydrodynamic microenvironment of osteocytes in microgravity. Comput Methods Biomech Biomed Engin 2022; 25:1757-1766. [PMID: 35170387 DOI: 10.1080/10255842.2022.2037130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Osteoporosis occurs in astronauts after long-term space flight owing to the lack of gravity. The mechanical microenvironment of osteocytes in load-bearing bone are changed during resistance exercise, which prevents massive bone loss in the human body. A cylindrical fluid-structure coupling finite element model for osteons with a two-stage pore structure (i.e., Haversian canal, lacunar-canalicular system) was established with the software COMSOL. In the Earth's gravity field and in microgravity, considering the effects of pulsating pressure of arterioles, a comparative study was performed on the changes in hydrodynamic microenvironment of osteocytes during human body high-intensity exercise at different frequencies (defined as causing bone to produce 3000 με) and the body is at rest. Positive and negative liquid pressure (with respect to one atmosphere pressure) alternately acted on osteocytes during human exercising, but only positive pressure acted on osteocytes during human resting. The variation range of liquid pressure acted on osteocytes during human exercising was significantly higher than that during resting. The liquid flow velocity around osteocytes during body exercise was about four orders of magnitude higher than that during resting. In microgravity, moderate physical exercise can obviously improve the hydrodynamic microenvironment of osteocytes in load-bearing bone, which could compensate for the lack of mechanical stimulation to osteocytes caused by the lack of gravity, thereby promoting the normal physiological function of osteocytes. To a certain extent, these results revealed the biomechanical mechanism by which exercise has an effect in fighting osteoporosis in astronauts.
Collapse
Affiliation(s)
- Hai-Ying Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, People's Republic of China.,National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, People's Republic of China
| | - Chao-Hui Zhao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, People's Republic of China.,National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, People's Republic of China
| | - Hao Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, People's Republic of China.,National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, People's Republic of China
| | - Wei Wang
- Department of Mechanics, School of Mechanical Engineering, Tianjin University, Tianjin, People's Republic of China
| | - Qing-Jian Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, People's Republic of China.,National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, People's Republic of China
| |
Collapse
|
27
|
Vittrup SØ, Hanberg P, Knudsen MB, Tøstesen SK, Kipp JO, Hansen J, Jørgensen NP, Stilling M, Bue M. Tibial bone and soft-tissue concentrations following combination therapy with vancomycin and meropenem - evaluated by microdialysis in a porcine model : should patients with open fractures have higher doses of antibiotics? Bone Joint Res 2022; 11:112-120. [PMID: 35176868 PMCID: PMC8882321 DOI: 10.1302/2046-3758.112.bjr-2021-0321.r1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aims Prompt and sufficient broad-spectrum empirical antibiotic treatment is key to preventing infection following open tibial fractures. Succeeding co-administration, we dynamically assessed the time for which vancomycin and meropenem concentrations were above relevant epidemiological cut-off (ECOFF) minimal inhibitory concentrations (T > MIC) in tibial compartments for the bacteria most frequently encountered in open fractures. Low and high MIC targets were applied: 1 and 4 µg/ml for vancomycin, and 0.125 and 2 µg/ml for meropenem. Methods Eight pigs received a single dose of 1,000 mg vancomycin and 1,000 mg meropenem simultaneously over 100 minutes and 10 minutes, respectively. Microdialysis catheters were placed for sampling over eight hours in tibial cancellous bone, cortical bone, and adjacent subcutaneous adipose tissue. Venous blood samples were collected as references. Results Across the targeted ECOFF values, vancomycin displayed longer T > MIC in all the investigated compartments in comparison to meropenem. For both drugs, cortical bone exhibited the shortest T > MIC. For the low MIC targets and across compartments, mean T > MIC ranged between 208 and 449 minutes (46% to 100%) for vancomycin and between 189 and 406 minutes (42% to 90%) for meropenem. For the high MIC targets, mean T > MIC ranged between 30 and 446 minutes (7% to 99%) for vancomycin and between 45 and 181 minutes (10% to 40%) for meropenem. Conclusion The differences in the T > MIC between the low and high targets illustrate how the interpretation of these results is highly susceptible to the defined MIC target. To encompass any trauma, contamination, or individual tissue differences, a more aggressive dosing approach may be considered to achieve longer T > MIC in all the exposed tissues, and thereby lower the risk of acquiring an infection after open tibial fractures. Cite this article: Bone Joint Res 2022;11(2):112–120.
Collapse
Affiliation(s)
- Sofus Ørbæk Vittrup
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Pelle Hanberg
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin Bruun Knudsen
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark
| | - Sara Kousgaard Tøstesen
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Josephine Olsen Kipp
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jakob Hansen
- Department of Forensic Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Maiken Stilling
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Mats Bue
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
28
|
Osteocytic Pericellular Matrix (PCM): Accelerated Degradation under In Vivo Loading and Unloading Conditions Using a Novel Imaging Approach. Genes (Basel) 2021; 13:genes13010072. [PMID: 35052411 PMCID: PMC8775093 DOI: 10.3390/genes13010072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 01/03/2023] Open
Abstract
The proteoglycan-containing pericellular matrix (PCM) controls both the biophysical and biochemical microenvironment of osteocytes, which are the most abundant cells embedded and dispersed in bones. As a molecular sieve, osteocytic PCMs not only regulate mass transport to and from osteocytes but also act as sensors of external mechanical environments. The turnover of osteocytic PCM remains largely unknown due to technical challenges. Here, we report a novel imaging technique based on metabolic labeling and “click-chemistry,” which labels de novo PCM as “halos” surrounding osteocytes in vitro and in vivo. We then tested the method and showed different labeling patterns in young vs. old bones. Further “pulse-chase” experiments revealed dramatic difference in the “half-life” of PCM of cultured osteocytes (~70 h) and that of osteocytes in vivo (~75 d). When mice were subjected to either 3-week hindlimb unloading or 7-week tibial loading (5.1 N, 4 Hz, 3 d/week), PCM half-life was shortened (~20 d) and degradation accelerated. Matrix metallopeptidase MMP-14 was elevated in mechanically loaded osteocytes, which may contribute to PCM degradation. This study provides a detailed procedure that enables semi-quantitative study of the osteocytic PCM remodeling in vivo and in vitro.
Collapse
|
29
|
ZHANG HAO, LIU HAIYING, ZHANG CHUNQIU, LIU ZHENZHONG, WANG WEI. MULTI-SCALE MECHANICAL BEHAVIOR ANALYSIS ON FLUID–SOLID COUPLING FOR OSTEONS IN VARIOUS GRAVITATIONAL FIELDS. J MECH MED BIOL 2021. [DOI: 10.1142/s0219519421500718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background: Compact bone mainly consists of cylindrical osteon structures. In microgravity, the change in the mechanical microenvironment of osteocytes might be the root cause of astronauts’ bone loss during space flights. Methods: A multi-scale three-dimensional (3D) fluid–solid coupling finite element model of osteons with a two-stage pore structure was developed using COMSOL software based on the natural structure of osteocytes. Gradients in gravitational fields of [Formula: see text]1, 0, 1, 2.5, and 3.7[Formula: see text]g were used to investigate the changes in the mechanical microenvironment on osteocyte structure. The difference in arteriole pulsating pressure and static compression stress caused by each gravity gradient was investigated. Results: The mechanical response of osteocytes increased with the value of g, compared with the Earth’s gravitational field. For instance, the fluid pressure of osteocytes and the von Mises stress of bone matrix near lacunae decreased by 31.3% and 99.9%, respectively, in microgravity. Under static loading, only about 16.7% of osteocytes in microgravity and 58.3% of osteocytes in the Earth’s gravitational field could reach the fluid shear stress threshold of biological reactions in cell culture experiments. Compared with the Earth’s gravitational field, the pressure gradient inside osteocytes severely decreased in microgravity. Conclusion: The mechanical microenvironment of osteocytes in microgravity might cause significant changes in the mechanical microenvironment of osteocytes, which may lead to disuse osteoporosis in astronauts.
Collapse
Affiliation(s)
- HAO ZHANG
- Tianjin Key Laboratory for Advanced Mechatronic, System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
- National Demonstration Center for Experimental, Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - HAI-YING LIU
- Tianjin Key Laboratory for Advanced Mechatronic, System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
- National Demonstration Center for Experimental, Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - CHUN-QIU ZHANG
- Tianjin Key Laboratory for Advanced Mechatronic, System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
- National Demonstration Center for Experimental, Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - ZHEN-ZHONG LIU
- Tianjin Key Laboratory for Advanced Mechatronic, System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, P. R. China
- National Demonstration Center for Experimental, Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - WEI WANG
- Department of Mechanics, School of Mechanical Engineering, Tianjin University, Tianjin 300354, P. R. China
| |
Collapse
|
30
|
Aguirre JI, Castillo EJ, Kimmel DB. Biologic and pathologic aspects of osteocytes in the setting of medication-related osteonecrosis of the jaw (MRONJ). Bone 2021; 153:116168. [PMID: 34487892 PMCID: PMC8478908 DOI: 10.1016/j.bone.2021.116168] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/02/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023]
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a potentially severe, debilitating condition affecting patients with cancer and patients with osteoporosis who have been treated with powerful antiresorptives (pARs) or angiogenesis inhibitors (AgIs). Oral risk factors associated with the development of MRONJ include tooth extraction and inflammatory dental disease (e.g., periodontitis, periapical infection). In bone tissues, osteocytes play a bidirectional role in which they not only act as the "receiver" of systemic signals from blood vessels, such as hormones and drugs, or local signals from the mineralized matrix as it is deformed, but they also play a critical role as "transmitter" of signals to the cells that execute bone modeling and remodeling (osteoclasts, osteoblasts and lining cells). When the survival capacity of osteocytes is overwhelmed, they can die. Osteocyte death has been associated with several pathological conditions. Whereas the causes and mechanisms of osteocyte death have been studied in conditions like osteonecrosis of the femoral head (ONFH), few studies of the causes and mechanisms of osteocyte death have been done in MRONJ. The three forms of cell death that affect most of the different cells in the body (apoptosis, autophagy, and necrosis) have been recognized in osteocytes. Notably, necroptosis, a form of regulated cell death with "a necrotic cell death phenotype," has also been identified as a form of cell death in osteocytes under certain pathologic conditions. Improving the understanding of osteocyte death in MRONJ may be critical for preventing disease and developing treatment approaches. In this review, we intend to provide insight into the biology of osteocytes, cell death, in general, and osteocyte death, in particular, and discuss hypothetical mechanisms involved in osteocyte death associated with MRONJ.
Collapse
Affiliation(s)
- J I Aguirre
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - E J Castillo
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - D B Kimmel
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America
| |
Collapse
|
31
|
Cen H, Yao Y, Liu H, Jia S, Gong H. Multiscale mechanical responses of young and elderly human femurs: A finite element investigation. Bone 2021; 153:116125. [PMID: 34280582 DOI: 10.1016/j.bone.2021.116125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Bone remodeling in the elderly is no longer balanced. As a result, the morphologies and mechanical properties of bone at different scales will change. These changes would affect the mechanical responses of bone, which might exacerbate the imbalance of bone remodeling and even cause age-related bone diseases. METHODS Considering those changes, multiscale finite element (FE) models of bone in the young and the elderly were developed that included macroscale (proximal femur), mesoscale (cortical bone), microscale (Haversian system) and sub-microscale (osteocyte-lacuna-canaliculus-extracellular matrix system, OLCES). The stress and strain distributions at different scales and transmissions among different scales were investigated. RESULTS The stresses of the elderly at macroscale, mesoscale and microscale were higher than those in the young by 23.7%, 62.5% and 8.0%, respectively, and the stresses of the elderly and the young at sub-microscale were almost the same. The strain of the elderly at macroscale, mesoscale, microscale and sub-microscale were higher than those in the young by 48.6%, 56.8%, 11.9% and 25.1%, respectively. The stress and strain transmission rates (ησand ηε) from mesoscale to microscale were decreased by 1.8%, and 2.5% than those from macroscale to mesoscale in the elderly, respectively; but increased by 13.8%, and 4.7% in the young, respectively. ηε from microscale to sub-microscale in the elderly was higher than that in the young by 21.3%. CONCLUSIONS Degeneration of cortical bone mechanical property in the elderly causes increases in stress and strain at macroscale and mesoscale. The reduction of lacunar number in the elderly is not conducive to the mechanical transmission from mesoscale to microscale. The differences in stress and strain at microscale between the young and the elderly are smaller than those at macroscale or mesoscale. The strain stimulus sensed by osteocyte in the elderly is not weakened compared with that in the young.
Collapse
Affiliation(s)
- Haipeng Cen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Yan Yao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Haibo Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Shaowei Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - He Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
32
|
Yokoyama Y, Kameo Y, Kamioka H, Adachi T. High-resolution image-based simulation reveals membrane strain concentration on osteocyte processes caused by tethering elements. Biomech Model Mechanobiol 2021; 20:2353-2360. [PMID: 34471950 PMCID: PMC8595188 DOI: 10.1007/s10237-021-01511-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/13/2021] [Indexed: 11/08/2022]
Abstract
Osteocytes are vital for regulating bone remodeling by sensing the flow-induced mechanical stimuli applied to their cell processes. In this mechanosensing mechanism, tethering elements (TEs) connecting the osteocyte process with the canalicular wall potentially amplify the strain on the osteocyte processes. The ultrastructure of the osteocyte processes and canaliculi can be visualized at a nanometer scale using high-resolution imaging via ultra-high voltage electron microscopy (UHVEM). Moreover, the irregular shapes of the osteocyte processes and the canaliculi, including the TEs in the canalicular space, should considerably influence the mechanical stimuli applied to the osteocytes. This study aims to characterize the roles of the ultrastructure of osteocyte processes and canaliculi in the mechanism of osteocyte mechanosensing. Thus, we constructed a high-resolution image-based model of an osteocyte process and a canaliculus using UHVEM tomography and investigated the distribution and magnitude of flow-induced local strain on the osteocyte process by performing fluid–structure interaction simulation. The analysis results reveal that local strain concentration in the osteocyte process was induced by a small number of TEs with high tension, which were inclined depending on the irregular shapes of osteocyte processes and canaliculi. Therefore, this study could provide meaningful insights into the effect of ultrastructure of osteocyte processes and canaliculi on the osteocyte mechanosensing mechanism.
Collapse
Affiliation(s)
- Yuka Yokoyama
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshitaka Kameo
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-ku, Okayama, 700-8525, Japan
| | - Taiji Adachi
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
33
|
Schurman CA, Verbruggen SW, Alliston T. Disrupted osteocyte connectivity and pericellular fluid flow in bone with aging and defective TGF-β signaling. Proc Natl Acad Sci U S A 2021; 118:e2023999118. [PMID: 34161267 PMCID: PMC8237574 DOI: 10.1073/pnas.2023999118] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Skeletal fragility in the elderly does not simply result from a loss of bone mass. However, the mechanisms underlying the concurrent decline in bone mass, quality, and mechanosensitivity with age remain unclear. The important role of osteocytes in these processes and the age-related degeneration of the intricate lacunocanalicular network (LCN) in which osteocytes reside point to a primary role for osteocytes in bone aging. Since LCN complexity severely limits experimental dissection of these mechanisms in vivo, we used two in silico approaches to test the hypothesis that LCN degeneration, due to aging or an osteocyte-intrinsic defect in transforming growth factor beta (TGF-β) signaling (TβRIIocy-/-), is sufficient to compromise essential osteocyte responsibilities of mass transport and exposure to mechanical stimuli. Using reconstructed confocal images of bone with fluorescently labeled osteocytes, we found that osteocytes from aged and TβRIIocy-/- mice had 33 to 45% fewer, and more tortuous, canaliculi. Connectomic network analysis revealed that diminished canalicular density is sufficient to impair diffusion even with intact osteocyte numbers and overall LCN architecture. Computational fluid dynamics predicts that the corresponding drop in shear stress experienced by aged or TβRIIocy-/- osteocytes is highly sensitive to canalicular surface area but not tortuosity. Simulated expansion of the osteocyte pericellular space to mimic osteocyte perilacunar/canalicular remodeling restored predicted shear stress for aged osteocytes to young levels. Overall, these models show how loss of LCN volume through LCN pruning may lead to impaired fluid dynamics and osteocyte exposure to mechanostimulation. Furthermore, osteocytes emerge as targets of age-related therapeutic efforts to restore bone health and function.
Collapse
Affiliation(s)
- Charles A Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA 94143
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94143
| | - Stefaan W Verbruggen
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom, E1 4NS
- Department of Mechanical Engineering, University of Sheffield, Sheffield, United Kingdom, S1 3JD
- The Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom, S1 3JD
- Department of Biomedical Engineering, Columbia University in the City of New York, New York, NY 10027
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA 94143;
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94143
| |
Collapse
|
34
|
Geoghegan IP, McNamara LM, Hoey DA. Estrogen withdrawal alters cytoskeletal and primary ciliary dynamics resulting in increased Hedgehog and osteoclastogenic paracrine signalling in osteocytes. Sci Rep 2021; 11:9272. [PMID: 33927279 PMCID: PMC8085225 DOI: 10.1038/s41598-021-88633-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023] Open
Abstract
Estrogen deficiency during post-menopausal osteoporosis leads to osteoclastogenesis and bone loss. Increased pro-osteoclastogenic signalling (RANKL/OPG) by osteocytes occurs following estrogen withdrawal (EW) and is associated with impaired focal adhesions (FAs) and a disrupted actin cytoskeleton. RANKL production is mediated by Hedgehog signalling in osteocytes, a signalling pathway associated with the primary cilium, and the ciliary structure is tightly coupled to the cytoskeleton. Therefore, the objective of this study was to investigate the role of the cilium and associated signalling in EW-mediated osteoclastogenic signalling in osteocytes. We report that EW leads to an elongation of the cilium and increase in Hedgehog and osteoclastogenic signalling. Significant trends were identified linking cilia elongation with reductions in cell area and % FA area/cell area, indicating that cilia elongation is associated with disruption of FAs and actin contractility. To verify this, we inhibited FA assembly via αvβ3 antagonism and inhibited actin contractility and demonstrated an elongated cilia and increased expression of Hh markers and Rankl expression. Therefore, our results suggest that the EW conditions associated with osteoporosis lead to a disorganisation of αvβ3 integrins and reduced actin contractility, which were associated with an elongation of the cilium, activation of the Hh pathway and osteoclastogenic paracrine signalling.
Collapse
Affiliation(s)
- Ivor P Geoghegan
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Science and Engineering, National University of Ireland, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Laoise M McNamara
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Science and Engineering, National University of Ireland, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - David A Hoey
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland. .,Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02 R590, Ireland. .,Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland. .,Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2, Ireland.
| |
Collapse
|
35
|
Gauthier R, Jeannin C, Attik N, Trunfio-Sfarghiu AM, Gritsch K, Grosgogeat B. Tissue Engineering for Periodontal Ligament Regeneration: Biomechanical Specifications. J Biomech Eng 2021; 143:030801. [PMID: 33067629 DOI: 10.1115/1.4048810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Indexed: 11/08/2022]
Abstract
The periodontal biomechanical environment is very difficult to investigate. By the complex geometry and composition of the periodontal ligament (PDL), its mechanical behavior is very dependent on the type of loading (compressive versus tensile loading; static versus cyclic loading; uniaxial versus multiaxial) and the location around the root (cervical, middle, or apical). These different aspects of the PDL make it difficult to develop a functional biomaterial to treat periodontal attachment due to periodontal diseases. This review aims to describe the structural and biomechanical properties of the PDL. Particular importance is placed in the close interrelationship that exists between structure and biomechanics: the PDL structural organization is specific to its biomechanical environment, and its biomechanical properties are specific to its structural arrangement. This balance between structure and biomechanics can be explained by a mechanosensitive periodontal cellular activity. These specifications have to be considered in the further tissue engineering strategies for the development of an efficient biomaterial for periodontal tissues regeneration.
Collapse
Affiliation(s)
- R Gauthier
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France
| | - Christophe Jeannin
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France; Hospices Civils de Lyon, Service d'Odontologie, Lyon 69007, France
| | - N Attik
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France
| | | | - K Gritsch
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France; Hospices Civils de Lyon, Service d'Odontologie, Lyon 69007, France
| | - B Grosgogeat
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France; Hospices Civils de Lyon, Service d'Odontologie, Lyon 69007, France
| |
Collapse
|
36
|
Malkovskiy AV, Van Wassenhove LD, Goltsev Y, Osei-Sarfo K, Chen CH, Efron B, Gudas LJ, Mochly-Rosen D, Rajadas J. The Effect of Ethanol Consumption on Composition and Morphology of Femur Cortical Bone in Wild-Type and ALDH2*2-Homozygous Mice. Calcif Tissue Int 2021; 108:265-276. [PMID: 33068139 PMCID: PMC8092984 DOI: 10.1007/s00223-020-00769-1] [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: 02/05/2020] [Accepted: 10/05/2020] [Indexed: 11/28/2022]
Abstract
ALDH2 inactivating mutation (ALDH2*2) is the most abundant mutation leading to bone morphological aberration. Osteoporosis has long been associated with changes in bone biomaterial in elderly populations. Such changes can be exacerbated with elevated ethanol consumption and in subjects with impaired ethanol metabolism, such as carriers of aldehyde dehydrogenase 2 (ALDH2)-deficient gene, ALDH2*2. So far, little is known about bone compositional changes besides a decrease in mineralization. Raman spectroscopic imaging has been utilized to study the changes in overall composition of C57BL/6 female femur bone sections, as well as in compound spatial distribution. Raman maps of bone sections were analyzed using multilinear regression with these four isolated components, resulting in maps of their relative distribution. A 15-week treatment of both wild-type (WT) and ALDH2*2/*2 mice with 20% ethanol in the drinking water resulted in a significantly lower mineral content (p < 0.05) in the bones. There was no significant change in mineral and collagen content due to the mutation alone (p > 0.4). Highly localized islets of elongated adipose tissue were observed on most maps. Elevated fat content was found in ALDH2*2 knock-in mice consuming ethanol (p < 0.0001) and this effect appeared cumulative. This work conclusively demonstrates that that osteocytes in femurs of older female mice accumulate fat, as has been previously theorized, and that fat accumulation is likely modulated by levels of acetaldehyde, the ethanol metabolite.
Collapse
Affiliation(s)
- Andrey V Malkovskiy
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford Medical School, Stanford, CA, 94305, USA.
- Department of Chemical and Systems Biology, Stanford Medical School, Stanford, CA, 94305, USA.
| | - Lauren D Van Wassenhove
- Department of Chemical and Systems Biology, Stanford Medical School, Stanford, CA, 94305, USA
| | - Yury Goltsev
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford Medical School, Stanford, CA, 94305, USA
| | - Kwame Osei-Sarfo
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford Medical School, Stanford, CA, 94305, USA
| | - Bradley Efron
- Department of Biomedical Data Science, Stanford Medical School, Stanford, CA, 94305, USA
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford Medical School, Stanford, CA, 94305, USA
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford Medical School, Stanford, CA, 94305, USA.
| |
Collapse
|
37
|
El-Gazzar A, Högler W. Mechanisms of Bone Fragility: From Osteogenesis Imperfecta to Secondary Osteoporosis. Int J Mol Sci 2021; 22:ijms22020625. [PMID: 33435159 PMCID: PMC7826666 DOI: 10.3390/ijms22020625] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Bone material strength is determined by several factors, such as bone mass, matrix composition, mineralization, architecture and shape. From a clinical perspective, bone fragility is classified as primary (i.e., genetic and rare) or secondary (i.e., acquired and common) osteoporosis. Understanding the mechanism of rare genetic bone fragility disorders not only advances medical knowledge on rare diseases, it may open doors for drug development for more common disorders (i.e., postmenopausal osteoporosis). In this review, we highlight the main disease mechanisms underlying the development of human bone fragility associated with low bone mass known to date. The pathways we focus on are type I collagen processing, WNT-signaling, TGF-ß signaling, the RANKL-RANK system and the osteocyte mechanosensing pathway. We demonstrate how the discovery of most of these pathways has led to targeted, pathway-specific treatments.
Collapse
Affiliation(s)
| | - Wolfgang Högler
- Correspondence: ; Tel.: +43-(0)5-7680-84-22001; Fax: +43-(0)5-7680-84-22004
| |
Collapse
|
38
|
Dole NS, Yee CS, Schurman CA, Dallas SL, Alliston T. Assessment of Osteocytes: Techniques for Studying Morphological and Molecular Changes Associated with Perilacunar/Canalicular Remodeling of the Bone Matrix. Methods Mol Biol 2021; 2230:303-323. [PMID: 33197021 PMCID: PMC9165628 DOI: 10.1007/978-1-0716-1028-2_17] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent advances have revived interest in the concept of osteocyte perilacunar/canalicular remodeling (PLR) and have motivated efforts to identify the mechanisms regulating this process in bone in the context of normal physiology and pathological conditions. Here, we describe several methods that are evaluating morphological changes associated with PLR function of osteocytes.
Collapse
Affiliation(s)
- Neha S Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Cristal S Yee
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Charles A Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri Kansas City, Kansas City, MO, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, USA.
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA, USA.
| |
Collapse
|
39
|
Zoller SD, Hegde V, Burke ZDC, Park HY, Ishmael CR, Blumstein GW, Sheppard W, Hamad C, Loftin AH, Johansen DO, Smith RA, Sprague MM, Hori KR, Clarkson SJ, Borthwell R, Simon SI, Miller JF, Nelson SD, Bernthal NM. Evading the host response: Staphylococcus "hiding" in cortical bone canalicular system causes increased bacterial burden. Bone Res 2020; 8:43. [PMID: 33303744 PMCID: PMC7728749 DOI: 10.1038/s41413-020-00118-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 08/22/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Extremity reconstruction surgery is increasingly performed rather than amputation for patients with large-segment pathologic bone loss. Debate persists as to the optimal void filler for this "limb salvage" surgery, whether metal or allograft bone. Clinicians focus on optimizing important functional gains for patients, and the risk of devastating implant infection has been thought to be similar regardless of implant material. Recent insights into infection pathophysiology are challenging this equipoise, however, with both basic science data suggesting a novel mechanism of infection of Staphylococcus aureus (the most common infecting agent) into the host lacunar-canaliculi network, and also clinical data revealing a higher rate of infection of allograft over metal. The current translational study was therefore developed to bridge the gap between these insights in a longitudinal murine model of infection of allograft bone and metal. Real-time Staphylococci infection characteristics were quantified in cortical bone vs metal, and both microarchitecture of host implant and presence of host immune response were assessed. An orders-of-magnitude higher bacterial burden was established in cortical allograft bone over both metal and cancellous bone. The establishment of immune-evading microabscesses was confirmed in both cortical allograft haversian canal and the submicron canaliculi network in an additional model of mouse femur bone infection. These study results reveal a mechanism by which Staphylococci evasion of host immunity is possible, contributing to elevated risks of infection in cortical bone. The presence of this local infection reservoir imparts massive clinical implications that may alter the current paradigm of osteomyelitis and bulk allograft infection treatment.
Collapse
Affiliation(s)
- Stephen D Zoller
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Vishal Hegde
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Zachary D C Burke
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Howard Y Park
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Chad R Ishmael
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Gideon W Blumstein
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - William Sheppard
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Christopher Hamad
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Amanda H Loftin
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Daniel O Johansen
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
| | - Ryan A Smith
- David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Marina M Sprague
- Department of Internal Medicine, University of California, Los Angeles, 757 Westwood Plaza, Suite 7501, Los Angeles, CA, 90095, USA
| | - Kellyn R Hori
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA
- David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Samuel J Clarkson
- David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Rachel Borthwell
- David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA
| | - Jeff F Miller
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, 90095, USA
| | - Scott D Nelson
- Department of Pathology, University of California, Los Angeles, 1250 16th St Suite 3450, Santa Monica, CA, 90404, USA
| | - Nicholas M Bernthal
- Department of Orthopedic Surgery, University of California, Los Angeles, 1250 16th St Suite 2100, Santa Monica, CA, 90404, USA.
| |
Collapse
|
40
|
Ellipsoidal mesoscale mineralization pattern in human cortical bone revealed in 3D by plasma focused ion beam serial sectioning. J Struct Biol 2020; 212:107615. [PMID: 32927057 DOI: 10.1016/j.jsb.2020.107615] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 01/24/2023]
Abstract
Visualizing bone mineralization and collagen fibril organization at intermediate scales between the nanometer and the hundreds of microns range, is still an important challenge. Similarly, visualizing cellular components which locally affect the tissue structure requires a precision of a few tens of nanometers at maximum while spanning several tens of micrometers. In the last decade, gallium focused ion beam (FIB) equipped with a scanning electron microscope (SEM) proved to be an extremely valuable structural tool to meet those ends. In this study, we assess the capability of a recent plasma FIB-SEM technology which provides a potential increase in measurement speed over gallium FIB-SEM, thus paving the way to larger volume analysis. Nanometer-scale layers of demineralized and mineralized unstained human femoral lamellar bone were sequentially sectioned over volumes of 6-16,000 μm3. Analysis of mineralized tissue revealed prolate ellipsoidal mineral clusters measuring approximately 1.1 µm in length by 700 nm at their maximum diameter. Those features, suggested by others in high resolution studies, appear here as a ubiquitous motif in mineralized lamellar bone over thousands of microns cubed, suggesting a heterogeneous and yet regular pattern of mineral deposition past the single collagen fibril level. This large scale view retained sufficient resolution to visualize the collagen fibrils while also partly visualizing the lacuno-canalicular network in three-dimensions. These findings are strong evidence for suitability of PFIB as a bone analysis tool and the need to revisit bone mineralization over multi-length scales with mineralized tissue.
Collapse
|
41
|
Ganesh T, Laughrey LE, Niroobakhsh M, Lara-Castillo N. Multiscale finite element modeling of mechanical strains and fluid flow in osteocyte lacunocanalicular system. Bone 2020; 137:115328. [PMID: 32201360 PMCID: PMC7354216 DOI: 10.1016/j.bone.2020.115328] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 12/20/2022]
Abstract
Osteocytes form over 90% of the bone cells and are postulated to be mechanosensors responsible for regulating the function of osteoclasts and osteoblasts in bone modeling and remodeling. Physical activity results in mechanical loading on the bones. Osteocytes are thought to be the main mechanosensory cells in bone. Upon load osteocytes secrete key factors initiating downstream signaling pathways that regulate skeletal metabolism including the Wnt/β-catenin signaling pathway. Osteocytes have dendritic structures and are housed in the lacunae and canaliculi within the bone matrix. Mechanical loading is known to have two primary effects, namely a mechanical strain (membrane disruption by stretching) on the lacunae/cells, and fluid flow, in the form of fluid flow shear stress (FFSS), in the space between the cell membranes and the lacuna-canalicular walls. In response, osteocytes get activated via a process called mechanotransduction in which mechanical signals are transduced to biological responses. The study of mechanotransduction is a complex subject involving principles of engineering mechanics as well as biological signaling pathway studies. Several length scales are involved as the mechanical loading on macro sized bones are converted to strain and FFSS responses at the micro-cellular level. Experimental measurements of strain and FFSS at the cellular level are very difficult and correlating them to specific biological activity makes this a very challenging task. One of the methods commonly adopted is a multi-scale approach that combines biological and mechanical experimentation with in silico numerical modeling of the engineering aspects of the problem. Finite element analysis along with fluid-structure interaction methodologies are used to compute the mechanical strain and FFSS. These types of analyses often involve a multi-length scale approach where models of both the macro bone structure and micro structure at the cellular length scale are used. Imaging modalities play a crucial role in the development of the models and present their own challenges. This paper reviews the efforts of various research groups in addressing this problem and presents the work in our research group. A clear understanding of how mechanical stimuli affect the lacunae and perilacunar tissue strains and shear stresses on the cellular membranes may ultimately lead to a better understanding of the process of osteocyte activation.
Collapse
Affiliation(s)
- Thiagarajan Ganesh
- Department of Civil and Mechanical Engineering, University of Missouri-Kansas City, 350L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO 64110, United States of America.
| | - Loretta E Laughrey
- Department of Civil and Mechanical Engineering, University of Missouri-Kansas City, 350L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO 64110, United States of America
| | - Mohammadmehdi Niroobakhsh
- Department of Civil and Mechanical Engineering, University of Missouri-Kansas City, 350L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO 64110, United States of America
| | - Nuria Lara-Castillo
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 650 E 25th Street, Kansas City, MO 64108, United States of America
| |
Collapse
|
42
|
Srivastava T, Joshi T, Jiang Y, Heruth DP, Rezaiekhaligh MH, Novak J, Staggs VS, Alon US, Garola RE, El-Meanawy A, McCarthy ET, Zhou J, Boinpelly VC, Sharma R, Savin VJ, Sharma M. Upregulated proteoglycan-related signaling pathways in fluid flow shear stress-treated podocytes. Am J Physiol Renal Physiol 2020; 319:F312-F322. [PMID: 32628542 DOI: 10.1152/ajprenal.00183.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The ultrafiltrate flow over the major processes and cell body generates fluid flow shear stress (FFSS) on podocytes. Hyperfiltration-associated increase in FFSS can lead to podocyte injury and detachment. Previously, we showed that FFSS-induced upregulation of the cyclooxygenase 2 (COX2)-PGE2-prostaglandin E receptor 2 (EP2) axis in podocytes activates Akt-glycogen synthase kinase-3β-β-catenin and MAPK/ERK signaling in response to FFSS. Integrative MultiOmics Pathway Resolution (IMPRes) is a new bioinformatic tool that enables simultaneous time-series analysis of more than two groups to identify pathways and molecular connections. In the present study, we used previously characterized COX2 [prostaglandin-endoperoxide synthase 2 (Ptgs2)], EP2 (Ptger2), and β1-catenin (Ctnnb1) as "seed genes" from an array data set of four groups analyzed over a time course. The 3 seed genes shared 7 pathways and 50 genes of 14 pathways and 89 genes identified by IMPRes. A composite of signaling pathways highlighted the temporal molecular connections during mechanotransduction signaling in FFSS-treated podocytes. We investigated the "proteoglycans in cancer" and "galactose metabolism" pathways predicted by IMPRes. A custom-designed PCR array validated 60.7% of the genes predicted by IMPRes analysis, including genes for the above-named pathways. Further validation using Western blot analysis showed increased expression of phosho-Erbb2, phospho-mammalian target of rapamycin (mTOR), CD44, and hexokinase II (Hk2); decreased total Erbb2, galactose mutarotase (Galm), and β-1,4-galactosyltransferase 1 (B4galt1); and unchanged total mTOR and AKT3. These findings corroborate our previously reported results. This study demonstrates the potential of the IMPRes method to identify novel pathways. Identifying the "proteoglycans in cancer" and "galactose metabolism" pathways has generated a lead to study the significance of FFSS-induced glycocalyx remodeling and possible detachment of podocytes from the glomerular matrix.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri.,Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Department of Oral and Craniofacial Sciences, University of Missouri School of Dentistry, Kansas City, Missouri
| | - Trupti Joshi
- Department of Health Management and Informatics and University of Missouri Informatics Institute, University of Missouri, Columbia, Missouri.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,MU Data Science and Informatics Institute, University of Missouri, Columbia, Missouri
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Daniel P Heruth
- Children's Mercy Research Institute, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Mohamed H Rezaiekhaligh
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Jan Novak
- Department of Microbiology, University of Alabama, Birmingham, Alabama
| | - Vincent S Staggs
- Biostatistics and Epidemiology Core, Children's Mercy Kansas City, Department of Pediatrics, University of Missouri, Kansas City, Missouri
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri, Kansas City, Missouri
| | - Robert E Garola
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital and University of Missouri, Kansas City
| | - Ashraf El-Meanawy
- Division of Nephrology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ellen T McCarthy
- Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Jianping Zhou
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Varun C Boinpelly
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Ram Sharma
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Virginia J Savin
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Mukut Sharma
- Midwest Veterans' Biomedical Research Foundation, Kansas City, Missouri.,Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| |
Collapse
|
43
|
McCutcheon S, Majeska RJ, Spray DC, Schaffler MB, Vazquez M. Apoptotic Osteocytes Induce RANKL Production in Bystanders via Purinergic Signaling and Activation of Pannexin Channels. J Bone Miner Res 2020; 35:966-977. [PMID: 31910292 PMCID: PMC8009310 DOI: 10.1002/jbmr.3954] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/25/2019] [Accepted: 12/14/2019] [Indexed: 12/15/2022]
Abstract
Localized apoptosis of osteocytes, the tissue-resident cells within bone, occurs with fatigue microdamage and activates bone resorption. Osteoclasts appear to target and remove dying osteocytes, resorbing damaged bone matrix as well. Osteocyte apoptosis similarly activates bone resorption with estrogen loss and in disuse. Apoptotic osteocytes trigger viable neighbor (ie, bystander) osteocytes to produce RANKL, the cytokine required for osteoclast activation. Signals from apoptotic osteocytes that trigger this bystander RANKL expression remain obscure. Studying signaling among osteocytes has been hampered by lack of in vitro systems that model the limited communication among osteocytes in vivo (ie, via gap junctions on cell processes and/or paracrine signals through thin pericellular fluid spaces around osteocytes). Here, we used a novel multiscale fluidic device (the Macro-micro-nano, or Mμn) that reproduces these key anatomical features. Osteocytes in discrete compartments of the device communicate only via these limited pathways, which allows assessment of their roles in triggering osteocytes RANKL expression. Apoptosis of MLOY-4 osteocytes in the Mμn device caused increased osteocyte RANKL expression in the neighboring compartment, consistent with in vivo findings. This RANKL upregulation in bystander osteocytes was prevented by blocking Pannexin 1 channels as well as its ATP receptor. ATP alone caused comparable RANKL upregulation in bystander osteocytes. Finally, blocking Connexin 43 gap junctions did not abolish osteocyte RANKL upregulation, but did alter the distribution of RANKL expressing bystander osteocytes. These findings point to extracellular ATP, released from apoptotic osteocytes via Panx1 channels, as a major signal for triggering bystander osteocyte RANKL expression and activating bone remodeling. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sean McCutcheon
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Robert J Majeska
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - David C Spray
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mitchell B Schaffler
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
44
|
Milovanovic P, Busse B. Phenomenon of osteocyte lacunar mineralization: indicator of former osteocyte death and a novel marker of impaired bone quality? Endocr Connect 2020; 9:R70-R80. [PMID: 32168472 PMCID: PMC7159263 DOI: 10.1530/ec-19-0531] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/13/2020] [Indexed: 11/09/2022]
Abstract
An increasing number of patients worldwide suffer from bone fractures that occur after low intensity trauma. Such fragility fractures are usually associated with advanced age and osteoporosis but also with long-term immobilization, corticosteroid therapy, diabetes mellitus, and other endocrine disorders. It is important to understand the skeletal origins of increased bone fragility in these conditions for preventive and therapeutic strategies to combat one of the most common health problems of the aged population. This review summarizes current knowledge pertaining to the phenomenon of micropetrosis (osteocyte lacunar mineralization). As an indicator of former osteocyte death, micropetrosis is more common in aged bone and osteoporotic bone. Considering that the number of mineralized osteocyte lacunae per bone area can distinguish healthy, untreated osteoporotic and bisphosphonate-treated osteoporotic patients, it could be regarded as a novel structural marker of impaired bone quality. Further research is needed to clarify the mechanism of lacunar mineralization and to explore whether it could be an additional target for preventing or treating bone fragility related to aging and various endocrine diseases.
Collapse
Affiliation(s)
- Petar Milovanovic
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Laboratory for Anthropology and Skeletal Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Correspondence should be addressed to B Busse:
| |
Collapse
|
45
|
Assessment of the human bone lacuno-canalicular network at the nanoscale and impact of spatial resolution. Sci Rep 2020; 10:4567. [PMID: 32165649 PMCID: PMC7067834 DOI: 10.1038/s41598-020-61269-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 02/17/2020] [Indexed: 11/09/2022] Open
Abstract
Recently, increasing attention has been given to the study of osteocytes, the cells that are thought to play an important role in bone remodeling and in the mechanisms of bone fragility. The interconnected osteocyte system is deeply embedded inside the mineralized bone matrix and lies within a closely fitted porosity known as the lacuno-canalicular network. However, quantitative data on human samples remain scarce, mostly measured in 2D, and there are gaps to be filled in terms of spatial resolution. In this work, we present data on femoral samples from female donors imaged with isotropic 3D spatial resolution by magnified X-ray phase nano computerized-tomography. We report quantitative results on the 3D structure of canaliculi in human femoral bone imaged with a voxel size of 30 nm. We found that the lacuno-canalicular porosity occupies on average 1.45% of the total tissue volume, the ratio of the canalicular versus lacunar porosity is about 37.7%, and the primary number of canaliculi stemming from each lacuna is 79 on average. The examination of this number at different distances from the surface of the lacunae demonstrates branching in the canaliculi network. We analyzed the impact of spatial resolution on quantification by comparing parameters extracted from the same samples imaged with 120 nm and 30 nm voxel sizes. To avoid any bias related to the analysis region, the volumes at 120 nm and 30 nm were registered and cropped to the same field of view. Our results show that the measurements at 120 and 30 nm are strongly correlated in our data set but that the highest spatial resolution provides more accurate information on the canaliculi network and its branching properties.
Collapse
|
46
|
Goggin P, Ho EML, Gnaegi H, Searle S, Oreffo ROC, Schneider P. Development of protocols for the first serial block-face scanning electron microscopy (SBF SEM) studies of bone tissue. Bone 2020; 131:115107. [PMID: 31669251 PMCID: PMC6961117 DOI: 10.1016/j.bone.2019.115107] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/27/2019] [Accepted: 10/09/2019] [Indexed: 11/28/2022]
Abstract
There is an unmet need for a high-resolution three-dimensional (3D) technique to simultaneously image osteocytes and the matrix in which these cells reside. In serial block-face scanning electron microscopy (SBF SEM), an ultramicrotome mounted within the vacuum chamber of a microscope repeatedly sections a resin-embedded block of tissue. Backscattered electron scans of the block face provide a stack of high-resolution two-dimensional images, which can be used to visualise and quantify cells and organelles in 3D. High-resolution 3D images of biological tissues from SBF SEM have been exploited considerably to date in the neuroscience field. However, non-brain samples, in particular hard biological tissues, have appeared more challenging to image by SBF SEM due to the difficulties of sectioning and rendering the samples conductive. We have developed and propose protocols for bone tissue preparation using SBF SEM, for imaging simultaneously soft and hard bone tissue components in 3D. We review the state of the art in high-resolution imaging of osteocytes, provide a historical perspective of SBF SEM, and we present first SBF SEM proof-of-concept studies for murine and human tissue. The application of SBF SEM to hard tissues will facilitate qualitative and quantitative 3D studies of tissue microstructure and ultrastructure in bone development, ageing and pathologies such as osteoporosis and osteoarthritis.
Collapse
Affiliation(s)
- Patricia Goggin
- Bioengineering Science Research Group, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Elaine M L Ho
- Bioengineering Science Research Group, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | | | | | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philipp Schneider
- Bioengineering Science Research Group, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
47
|
Pei S, Parthasarathy S, Parajuli A, Martinez J, Lv M, Jiang S, Wu D, Wei S, Lu XL, Farach-Carson MC, Kirn-Safran CB, Wang L. Perlecan/Hspg2 deficiency impairs bone's calcium signaling and associated transcriptome in response to mechanical loading. Bone 2020; 131:115078. [PMID: 31715337 PMCID: PMC6945981 DOI: 10.1016/j.bone.2019.115078] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 10/25/2022]
Abstract
Perlecan, a heparan sulfate proteoglycan, acts as a mechanical sensor for bone to detect external loading. Deficiency of perlecan increases the risk of osteoporosis in patients with Schwartz-Jampel Syndrome (SJS) and attenuates loading-induced bone formation in perlecan deficient mice (Hypo). Considering that intracellular calcium [Ca2+]i is an ubiquitous messenger controlling numerous cellular processes including mechanotransduction, we hypothesized that perlecan deficiency impairs bone's calcium signaling in response to loading. To test this, we performed real-time [Ca2+]i imaging on in situ osteocytes of adult murine tibiae under cyclic loading (8N). Relative to wild type (WT), Hypo osteocytes showed decreases in the overall [Ca2+]i response rate (-58%), calcium peaks (-33%), cells with multiple peaks (-53%), peak magnitude (-6.8%), and recovery speed to baseline (-23%). RNA sequencing and pathway analysis of tibiae from mice subjected to one or seven days of unilateral loading demonstrated that perlecan deficiency significantly suppressed the calcium signaling, ECM-receptor interaction, and focal adhesion pathways following repetitive loading. Defects in the endoplasmic reticulum (ER) calcium cycling regulators such as Ryr1/ryanodine receptors and Atp2a1/Serca1 calcium pumps were identified in Hypo bones. Taken together, impaired calcium signaling may contribute to bone's reduced anabolic response to loading, underlying the osteoporosis risk for the SJS patients.
Collapse
Affiliation(s)
- Shaopeng Pei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | | | - Ashutosh Parajuli
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Jerahme Martinez
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Mengxi Lv
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Sida Jiang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center, Houston, TX 77054, United States
| | - Shuo Wei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - X Lucas Lu
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center, Houston, TX 77054, United States
| | - Catherine B Kirn-Safran
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States; Department of Biology, Widener University, Chester, PA 19013, United States
| | - Liyun Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States; Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States; Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
48
|
Creecy A, Damrath JG, Wallace JM. Control of Bone Matrix Properties by Osteocytes. Front Endocrinol (Lausanne) 2020; 11:578477. [PMID: 33537002 PMCID: PMC7848033 DOI: 10.3389/fendo.2020.578477] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Osteocytes make up 90-95% of the cellular content of bone and form a rich dendritic network with a vastly greater surface area than either osteoblasts or osteoclasts. Osteocytes are well positioned to play a role in bone homeostasis by interacting directly with the matrix; however, the ability for these cells to modify bone matrix remains incompletely understood. With techniques for examining the nano- and microstructure of bone matrix components including hydroxyapatite and type I collagen becoming more widespread, there is great potential to uncover novel roles for the osteocyte in maintaining bone quality. In this review, we begin with an overview of osteocyte biology and the lacunar-canalicular system. Next, we describe recent findings from in vitro models of osteocytes, focusing on the transitions in cellular phenotype as they mature. Finally, we describe historical and current research on matrix alteration by osteocytes in vivo, focusing on the exciting potential for osteocytes to directly form, degrade, and modify the mineral and collagen in their surrounding matrix.
Collapse
Affiliation(s)
- Amy Creecy
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
| | - John G. Damrath
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
- *Correspondence: Joseph M. Wallace,
| |
Collapse
|
49
|
Liu XS, Wang L, de Bakker CMJ, Lai X. Mechanical Regulation of the Maternal Skeleton during Reproduction and Lactation. Curr Osteoporos Rep 2019; 17:375-386. [PMID: 31755029 PMCID: PMC7373497 DOI: 10.1007/s11914-019-00555-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review summarizes recently published data on the effects of pregnancy and lactation on bone structure, mechanical properties, and mechano-responsiveness in an effort to elucidate how the balance between the structural and metabolic functions of the skeleton is achieved during these physiological processes. RECENT FINDINGS While pregnancy and lactation induce significant changes in bone density and structure to provide calcium for fetal/infant growth, the maternal physiology also comprises several innate compensatory mechanisms that allow for the maintenance of skeletal mechanical integrity. Both clinical and animal studies suggest that pregnancy and lactation lead to adaptations in cortical bone structure to allow for rapid calcium release from the trabecular compartment while maintaining whole bone stiffness and strength. Moreover, extents of lactation-induced bone loss and weaning-induced recovery are highly dependent on a given bone's load-bearing function, resulting in better protection of the mechanical integrity at critical load-bearing sites. The recent discovery of lactation-induced osteocytic perilacunar/canalicular remodeling (PLR) indicates a new means for osteocytes to modulate mineral homeostasis and tissue-level mechanical properties of the maternal skeleton. Furthermore, lactation-induced PLR may also play an important role in maintaining the maternal skeleton's load-bearing capacity by altering osteocyte's microenvironment and modulating the transmission of anabolic mechanical signals to osteocytes. Both clinical and animal studies show that parity and lactation have no adverse, or a positive effect on bone strength later in life. The skeletal effects during pregnancy and lactation reflect an optimized balance between the mechanical and metabolic functions of the skeleton.
Collapse
Affiliation(s)
- X Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 332A Stemmler Hall, 36th Street and Hamilton Walk, Philadelphia, PA, USA.
| | - Liyun Wang
- Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Chantal M J de Bakker
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Xiaohan Lai
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
50
|
Wittig NK, Palle J, Østergaard M, Frølich S, Birkbak ME, Spiers KM, Garrevoet J, Birkedal H. Bone Biomineral Properties Vary across Human Osteonal Bone. ACS NANO 2019; 13:12949-12956. [PMID: 31613594 DOI: 10.1021/acsnano.9b05535] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The biomineralization of bone remains a puzzle. During Haversian remodeling in the dense human cortical bone, osteoclasts excavate a tunnel that is then filled in by osteoblasts with layers of bone of varying fibril orientations, resulting in a lamellar motif. Such bone represents an excellent possibility to increase our understanding of bone as a material as well as bone biomineralization by studying spatio/temporal variations in the biomineral across an osteon. To this end, fluorescence computed tomography and diffraction scattering computed tomography with sub-micrometer resolution is applied to obtain position resolved fluorescence spectra and diffraction patterns in a 3D volume. The microstructural properties of the apatite biomineral are not homogeneous but depend critically on the time point at which it was laid down. This indicates that the nature of bone biomineral is highly dependent on the microenvironment during bone formation and remodeling.
Collapse
Affiliation(s)
- Nina K Wittig
- Department of Chemistry and iNANO , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| | - Jonas Palle
- Department of Chemistry and iNANO , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| | - Maja Østergaard
- Department of Chemistry and iNANO , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| | - Simon Frølich
- Department of Chemistry and iNANO , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| | - Mie E Birkbak
- Department of Chemistry and iNANO , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| | | | - Jan Garrevoet
- DESY Photon Science , Notkestr. 85 , D-22607 Hamburg , Germany
| | - Henrik Birkedal
- Department of Chemistry and iNANO , Aarhus University , Gustav Wieds Vej 14 , 8000 Aarhus C , Denmark
| |
Collapse
|