1
|
Machireddy M, Nano S, Debiase L, Lewis J, Cole S, Li J, Niebur GL. Differential gene expression in trabecular bone osteocytes is related to the local strain and strain gradient. Sci Rep 2025; 15:18501. [PMID: 40425655 PMCID: PMC12117145 DOI: 10.1038/s41598-025-00214-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Osteocytes regulate the response of osteoclasts and osteoblasts to mechanical loading through signaling molecules, the levels of which are controlled by post-translational modification or degradation and by differential gene transcription and translation. The magnitude and mode of bone tissue deformation that elicits a transcriptional response in individual osteocytes in situ has been difficult to quantify. We measured SOST, Wnt11, TNF, and FRZB gene expression in osteocytes within loaded and unloaded control porcine trabecular bone explants using RNAScope® and compared the local tissue level strain and strain gradient-which we used as an indicator of potential poroelastic fluid flow-in the tissue surrounding osteocytes with high vs. low gene expression. The measured expression of all four genes differed between loaded and unloaded explants, on average, with the mean SOST expression level decreasing by 45%. In the loaded explants, gene expression was altered from baseline in about 30% of the osteocytes, and they were surrounded by tissue with higher strain and strain gradient than the 20 to 25% of osteocytes that remained near baseline expression. Both deviatoric strain and hydrostatic strain gradient were sensitive and specific predictors of the mechanobiological response for individual genes as well as combinations. SOST expression was highly related to elevated strain gradient, providing evidence that osteocytes respond to fluid flow in the lacunar-canalicular system.
Collapse
Affiliation(s)
- Meghana Machireddy
- University of Notre Dame, Bioengineering Graduate Program, Notre Dame, IN, USA
| | - Sarah Nano
- University of Notre Dame, Bioengineering Graduate Program, Notre Dame, IN, USA
| | - Lucas Debiase
- Department of Aerospace and Mechanical Engineering, Notre Dame, IN, USA
| | - Jonathan Lewis
- Department of Aerospace and Mechanical Engineering, Notre Dame, IN, USA
| | - Sara Cole
- Notre Dame Integrated Imaging Facility, Notre Dame, IN, USA
| | - Jun Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, USA
| | - Glen L Niebur
- University of Notre Dame, Bioengineering Graduate Program, Notre Dame, IN, USA.
- Department of Aerospace and Mechanical Engineering, Notre Dame, IN, USA.
| |
Collapse
|
2
|
Gao Y, Zhao S, Yang A. Numerical Simulation of Fluid Shear Stress Distribution in Microcracks of Trabecular Bone. Appl Bionics Biomech 2025; 2025:5634808. [PMID: 39850532 PMCID: PMC11753853 DOI: 10.1155/abb/5634808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Bone is one of the hardest tissues in the human body, but it can undergo microcracks under long-term and periodic mechanical loads. The Newton iterative method was used to calculate the steady state, and the effects of different inlet and outlet pressures, trabecular gap width and height, and microcrack's depth and width on the fluid shear stress (FSS) were studied, and the gradient of FSS inside the microcrack was analyzed. The results show that the pressure difference and trabecular gap heigh are positively correlated with the FSS (the linear correlation coefficients R 2 were 0.9768 and 0.96542, respectively). When the trabecular gap width was 100 μm, the peak of FSS decreased by 28.57% compared with 800 and 400 μm, and the gradient of FSS inside the microcrack was 0.1-0.4 Pa/mm. This study can help people more intuitively understand the internal fluid distribution of trabecular bone and provide a reliable theoretical basis for the subsequent construction of gradient FSS devices in vitro.
Collapse
Affiliation(s)
- Yan Gao
- Capital University of Physical Education and Sports, Institute of Artificial Intelligence in Sports, Beijing 100191, China
| | - Sen Zhao
- Beijing Institute of Technology, School of Aerospace Engineering, Beijing 100081, China
| | - Ailing Yang
- Beijing Institute of Technology, School of Aerospace Engineering, Beijing 100081, China
| |
Collapse
|
3
|
Gupta A, Saha S, Das A, Roy Chowdhury A. Evaluating the influence on osteocyte mechanobiology within the lacunar-canalicular system for varying lacunar equancy and perilacunar elasticity: A multiscale fluid-structure interaction analysis. J Mech Behav Biomed Mater 2024; 160:106767. [PMID: 39393133 DOI: 10.1016/j.jmbbm.2024.106767] [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: 06/07/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
The lacunar morphology and perilacunar tissue properties of osteocytes in bone can vary under different physiological and pathological conditions. How these alterations collectively change the overall micromechanics of osteocytes in the lacunar-canalicular system (LCS) of an osteon still requires special focus. Therefore, a Haversian canal and LCS-based osteon model was established to evaluate the changes in the hydrodynamic environment around osteocytes under physiological loading using fluid-structure interaction analysis, followed by a sub-modelled finite element analysis to assess the mechanical responses of osteocytes and their components. Osteocytes were modelled with detailed configurations, including cytoplasm, nucleus, and cytoskeleton, and parametric variations in lacunar equancy (L.Eq) and perilacunar elasticity (Pl.E) were considered within the osteon model. The study aimed to conduct a comparative study among osteon models with varying L. Eq and Pl. E to check the resulting differences in osteocyte mechanobiology. The results demonstrated that the average mechanical stimulation of each subcellular component of osteocytes increased with decreases in L. Eq and Pl. E, reflecting conditions typically seen in young, healthy bone as per previous literature. However, hydrodynamic responses, such as fluid flow and fluid shear stress on osteocytes, varied proportionally with the elasticity difference between the bone matrix and the perilacunar region during Pl. E variation. Additionally, the findings revealed that a minimal percentage of energy was used to transmit mechanical responses through microtubules from the cell membrane to the nucleus, and this energy percentage increased with higher L. Eq. The outcomes of the study could help to quantify how the osteocyte microenvironment and its mechanosensitivity within cortical bone changes with L. Eq and Pl. E alterations in different bone conditions, from young to aged and healthy to diseased.
Collapse
Affiliation(s)
- Abhisek Gupta
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India
| | - Subrata Saha
- Department of Restorative Dentistry, University of Washington, Seattle, WA, USA
| | - Apurba Das
- Department of Mechanical Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India
| | - Amit Roy Chowdhury
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India.
| |
Collapse
|
4
|
Hagan ML, Tuladhar A, Yu K, Alhamad DW, Bensreti H, Dorn J, Piedra VM, Cantu N, Stokes EG, Blumenthal D, Roberts RL, Balayan V, Bass SM, Dickerson T, Cartelle AL, Montesinos-Cartagena M, Awad ME, Castro AA, Garland T, Cooley MA, Johnson M, Hamrick MW, McNeil PL, McGee-Lawrence ME. Osteocyte Sptbn1 Deficiency Alters Cell Survival and Mechanotransduction Following Formation of Plasma Membrane Disruptions (PMD) from Mechanical Loading. Calcif Tissue Int 2024; 115:725-743. [PMID: 39276238 DOI: 10.1007/s00223-024-01285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024]
Abstract
We and others have shown that application of high-level mechanical loading promotes the formation of transient plasma membrane disruptions (PMD) which initiate mechanotransduction. We hypothesized that increasing osteocyte cell membrane fragility, by disrupting the cytoskeleton-associated protein β2-spectrin (Sptbn1), could alter osteocytic responses and bone adaptation to loading in a PMD-related fashion. In MLO-Y4 cells, treatment with the spectrin-disrupting agent diamide or knockdown of Sptbn1 via siRNA increased the number of PMD formed by fluid shear stress. Primary osteocytes from an osteocyte-targeted DMP1-Cre Sptbn1 conditional knockout (CKO) model mimicked trends seen with diamide and siRNA treatment and suggested the creation of larger PMD, which repaired more slowly, for a given level of stimulus. Post-wounding cell survival was impaired in all three models, and calcium signaling responses from the wounded osteocyte were mildly altered in Sptbn1 CKO cultures. Although Sptbn1 CKO mice did not demonstrate an altered skeletal phenotype as compared to WT littermates under baseline conditions, they showed a blunted increase in cortical thickness when subjected to an osteogenic tibial loading protocol as well as evidence of increased osteocyte death (increased lacunar vacancy) in the loaded limb after 2 weeks of loading. The impaired post-wounding cell viability and impaired bone adaptation seen with Sptbn1 disruption support the existence of an important role for Sptbn1, and PMD formation, in osteocyte mechanotransduction and bone adaptation to mechanical loading.
Collapse
Affiliation(s)
- Mackenzie L Hagan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Anik Tuladhar
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Dima W Alhamad
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Husam Bensreti
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Jennifer Dorn
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Victor M Piedra
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Nicholas Cantu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Eric G Stokes
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Daniel Blumenthal
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Rachel L Roberts
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Vanshika Balayan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Sarah M Bass
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Thomas Dickerson
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Anabel Liyen Cartelle
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Marlian Montesinos-Cartagena
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Mohamed E Awad
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Alberto A Castro
- Evolution Ecology & Organismal Biology Department, University of California Riverside, Riverside, USA
| | - Theodore Garland
- Evolution Ecology & Organismal Biology Department, University of California Riverside, Riverside, USA
| | - Marion A Cooley
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Maribeth Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Paul L McNeil
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, CB1101, Augusta, GA, 30912, USA.
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
5
|
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
|
6
|
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]
|
7
|
Sefa S, Espiritu J, Ćwieka H, Greving I, Flenner S, Will O, Beuer S, Wieland DF, Willumeit-Römer R, Zeller-Plumhoff B. Multiscale morphological analysis of bone microarchitecture around Mg-10Gd implants. Bioact Mater 2023; 30:154-168. [PMID: 37575877 PMCID: PMC10412723 DOI: 10.1016/j.bioactmat.2023.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
The utilization of biodegradable magnesium (Mg)-based implants for restoration of bone function following trauma represents a transformative approach in orthopaedic application. One such alloy, magnesium-10 weight percent gadolinium (Mg-10Gd), has been specifically developed to address the rapid degradation of Mg while enhancing its mechanical properties to promote bone healing. Previous studies have demonstrated that Mg-10Gd exhibits favorable osseointegration; however, it exhibits distinct ultrastructural adaptation in comparison to conventional implants like titanium (Ti). A crucial aspect that remains unexplored is the impact of Mg-10Gd degradation on the bone microarchitecture. To address this, we employed hierarchical three-dimensional imaging using synchrotron radiation in conjunction with image-based finite element modelling. By using the methods outlined, the vascular porosity, lacunar porosity and the lacunar-canaliculi network (LCN) morphology of bone around Mg-10Gd in comparison to Ti in a rat model from 4 weeks to 20 weeks post-implantation was investigated. Our investigation revealed that within our observation period, the degradation of Mg-10Gd implants was associated with significantly lower (p < 0.05) lacunar density in the surrounding bone, compared to Ti. Remarkably, the LCN morphology and the fluid flow analysis did not significantly differ for both implant types. In summary, a more pronounced lower lacunae distribution rather than their morphological changes was detected in the surrounding bone upon the degradation of Mg-10Gd implants. This implies potential disparities in bone remodelling rates when compared to Ti implants. Our findings shed light on the intricate relationship between Mg-10Gd degradation and bone microarchitecture, contributing to a deeper understanding of the implications for successful osseointegration.
Collapse
Affiliation(s)
- Sandra Sefa
- Institute of Metallic Biomaterials, Helmholtz Zentrum Hereon, Geesthacht, Germany
| | | | - Hanna Ćwieka
- Institute of Metallic Biomaterials, Helmholtz Zentrum Hereon, Geesthacht, Germany
| | - Imke Greving
- Institute of Materials Physics, Helmholtz Zentrum Hereon, Geesthacht, Germany
| | - Silja Flenner
- Institute of Materials Physics, Helmholtz Zentrum Hereon, Geesthacht, Germany
| | - Olga Will
- Molecular Imaging North Competence Center, Kiel University, Kiel, Germany
| | - Susanne Beuer
- Fraunhofer Institut für Integrierte Systeme und Bauelementetechnologie (IISB), Erlangen, Germany
| | - D.C Florian Wieland
- Institute of Metallic Biomaterials, Helmholtz Zentrum Hereon, Geesthacht, Germany
| | | | | |
Collapse
|
8
|
Zhao C, Liu H, Tian C, Zhang C, Wang W. Multi-scale numerical simulation on mechano-transduction of osteocytes in different gravity fields. Comput Methods Biomech Biomed Engin 2023; 26:1419-1430. [PMID: 36048419 DOI: 10.1080/10255842.2022.2117552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/13/2022] [Accepted: 08/23/2022] [Indexed: 11/03/2022]
Abstract
A three-dimensional model for osteocyte was established to research the mechanisms of mechano-transduction and amplification of primary cilium and osteocyte process in every gravity field. The results showed that significant stress concentration was observed in the area of physical connection between TES and the osteocyte process, where the fluid shear stress (FSS) was around two orders of magnitude higher than that in other areas. Due to the significant amplification effect of the TES structure on mechanical stimulation, making osteocyte process the "optimal mechanical receptor". In microgravity, the mechanical signal conduction ability of the osteocyte decreased significantly.. HighlightsAt the micro-nano scale, a 3D model for single bone lacunae-osteocyte system is established.The stress amplification mechanism of the transverse element is verified.Compared with the primary cilium, osteocyte process is the 'optimal mechanical receptor'.In microgravity, the mechanical signal conduction ability of osteocyte system decreased.
Collapse
Affiliation(s)
- Chaohui 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
| | - Haiying 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
| | - Congbiao Tian
- 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
| | - Chunqiu 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
| |
Collapse
|
9
|
Boucetta A, Ramtani S, Garzón-Alvarado DA. Both network architecture and micro cracks effects on lacuno-canalicular liquid flow efficiency within the context of multiphysics approach for bone remodeling. J Mech Behav Biomed Mater 2023; 141:105780. [PMID: 36989871 DOI: 10.1016/j.jmbbm.2023.105780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/27/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023]
Abstract
When physical forces are applied to bone, its mechanical adaptive behaviors change according to the microarchitecture configuration. This leads to changes in biological and physical thresholds in the remodeling cell population, involving sensor cells (osteocytes) interacting with each other and changes in osteocyte shape due to variation in lacunar shape. The resulting alterations in fluid flow leads to changes in the membrane electrical potential and shear stress. Eventual creation of microcracks, may lead in turn to modify cell activity. In contrast, the redundancy in the lacuno canalicular network (LCN) interconnectivity maintains partial flow. Our goal was to investigate the role of fluid flow in LCN by proposing a model of electro-mechanical energy spread through inhomogeneous microarchitectures. We focused on mechano-sensitivity to changes in load-induced flow impacted by neighboring micro cracks and quantifying its critical role in changing, velocity, shear stress and orientation of liquid mass transportation from one cell to another. To enhance the concept of intricacy LCN micro-structure to fluid flow, we provide a new combined effects factor considered as osteocytes sensor efficiency. We customized an influence function for each osteocyte, coupling: in one hand, the spatial distribution within remodeling influence areas, conducting a significant fluid spread, leading hydro-dynamic behavior and impacted further by presence of micro cracks and; in other hand, the fluid electro kinetic behavior. As an attempt to fill the limitations stated by many of the recent studies, we reveal in numerical simulation, some results which cannot be measured in vitro/in vivo studies. Numerical calculations were performed in order to evaluate, among many others, how liquid flow conditions changes between lacunas, how the orientation and the magnitude of the governing flow in LCN can regulate osteocytes efficiency. In addition to be regulated by osteocytes, a direct effects of fluid flow are also acting on osteoblast activity. In summary, this new approach considers mechano-sensitivity in relation to liquid flow dynamic and suggests additional pathway for Osseo integration via osteoblast regulation. However, this novel modeling approach may help improve the mapping and design bone scaffolds and/or selection of scaffold implantation regions.
Collapse
Affiliation(s)
- Abdelkader Boucetta
- Université Sorbonne Paris Nord, CSPBA-LBPS, UMR CNRS 7244, Inst Galilee, 99 Ave JB Clement, Villetaneuse, France; GE VERNOVA, SS&O-OPS-O&M EMEA Regions, Algiers, Algeria.
| | - Salah Ramtani
- Université Sorbonne Paris Nord, CSPBA-LBPS, UMR CNRS 7244, Inst Galilee, 99 Ave JB Clement, Villetaneuse, France.
| | - Diego A Garzón-Alvarado
- Universidad Nacional de Colombia, Biomimetics Laboratory-Biotechnology Institute, Bogota, 571, Republic of Colombia.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Leser JM, Harriot A, Buck HV, Ward CW, Stains JP. Aging, Osteo-Sarcopenia, and Musculoskeletal Mechano-Transduction. FRONTIERS IN REHABILITATION SCIENCES 2021; 2:782848. [PMID: 36004321 PMCID: PMC9396756 DOI: 10.3389/fresc.2021.782848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022]
Abstract
The decline in the mass and function of bone and muscle is an inevitable consequence of healthy aging with early onset and accelerated decline in those with chronic disease. Termed osteo-sarcopenia, this condition predisposes the decreased activity, falls, low-energy fractures, and increased risk of co-morbid disease that leads to musculoskeletal frailty. The biology of osteo-sarcopenia is most understood in the context of systemic neuro-endocrine and immune/inflammatory alterations that drive inflammation, oxidative stress, reduced autophagy, and cellular senescence in the bone and muscle. Here we integrate these concepts to our growing understanding of how bone and muscle senses, responds and adapts to mechanical load. We propose that age-related alterations in cytoskeletal mechanics alter load-sensing and mechano-transduction in bone osteocytes and muscle fibers which underscores osteo-sarcopenia. Lastly, we examine the evidence for exercise as an effective countermeasure to osteo-sarcopenia.
Collapse
Affiliation(s)
| | | | | | | | - Joseph P. Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
Kameo Y, Ozasa M, Adachi T. Computational framework for analyzing flow-induced strain on osteocyte as modulated by microenvironment. J Mech Behav Biomed Mater 2021; 126:105027. [PMID: 34920322 DOI: 10.1016/j.jmbbm.2021.105027] [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: 09/14/2021] [Revised: 11/19/2021] [Accepted: 12/02/2021] [Indexed: 12/26/2022]
Abstract
Osteocytes buried in bone matrix are major mechanosensory cells that regulate bone remodeling in response to interstitial fluid flow in a lacuno-canalicular porosity. To gain an understanding of the mechanism of osteocyte mechanosensing, it is important to be able to evaluate the local strain on the osteocyte process membrane induced by the interstitial fluid flow. The microenvironment of the osteocytes, including the pericellular matrix (PCM) and canalicular ultrastructure, is a key modulator of the flow-induced strain on the osteocyte process membrane because it produces heterogeneous flow patterns in the pericellular space. To investigate the effect of changes in the microenvironment of osteocytes on the flow-induced strain, we developed a novel computational framework for analyzing the fluid-structure interaction. Computer simulations based on the proposed framework enabled evaluation of the spatial distribution of flow-induced strain on the osteocyte process membrane according to changes in the PCM density and canalicular curvature. The simulation results reveal that a decrease in PCM density and an increase in canalicular curvature, each of which is associated with aging and bone disease, have the notable effect of enhancing local flow-induced strain on the osteocyte process membrane. We believe that the proposed computational framework is a promising framework for investigating cell-specific mechanical stimuli and that it has the potential to accelerate the mechanobiological study of osteocytes by providing a deeper understanding of their mechanical environment in living bone tissue.
Collapse
Affiliation(s)
- Yoshitaka Kameo
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masahiro Ozasa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taiji Adachi
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
14
|
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: 55] [Impact Index Per Article: 13.8] [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
|
15
|
Smit TH. Closing the osteon: Do osteocytes sense strain rate rather than fluid flow? Bioessays 2021; 43:e2000327. [PMID: 34111316 DOI: 10.1002/bies.202000327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/05/2022]
Abstract
Osteons are cylindrical structures of bone created by matrix resorbing osteoclasts, followed by osteoblasts that deposit new bone. Osteons align with the principal loading direction and it is thought that the osteoclasts are directed by osteocytes, the mechanosensitive cells that reside inside the bone matrix. These osteocytes are presumably controlled by interstitial fluid flow, induced by the physiological loading of bones. Here I consider the stimulation of osteocytes while the osteon is closed by osteoblasts. In a conceptual finite element model, bone is considered a poro-elastic material and subjected to locomotion-induced loading conditions. It appears that the magnitude of flow is constant along the closing cone, while shear strain rate in the bone matrix diminishes linearly with the deposition of bone. This suggests that shear strain rate, rather than fluid flow, is the physical cue that controls osteocytes and bone deposition in newly formed osteons.
Collapse
Affiliation(s)
- Theodoor H Smit
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Department of Orthopaedic Surgery, Amsterdam University Medical Centers, Amsterdam Movement Sciences Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Gatti V, Gelbs MJ, Guerra RB, Gerber MB, Fritton SP. Interstitial fluid velocity is decreased around cortical bone vascular pores and depends on osteocyte position in a rat model of disuse osteoporosis. Biomech Model Mechanobiol 2021; 20:1135-1146. [PMID: 33666792 DOI: 10.1007/s10237-021-01438-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 02/15/2021] [Indexed: 10/22/2022]
Abstract
Muscle paralysis induced with botulinum toxin (Botox) injection increases vascular porosity and reduces osteocyte lacunar density in the tibial cortical bone of skeletally mature rats. These morphological changes potentially affect interstitial fluid flow in the lacunar-canalicular porosity, which is thought to play a role in osteocyte mechanotransduction. The aim of this study was to investigate the effects of disuse-induced morphological changes on interstitial fluid velocity around osteocytes in the bone cortex. Micro-CT images from a previous study that quantified the effects of Botox-induced muscle paralysis on bone microarchitecture in skeletally mature rats were used to create high-resolution, animal-specific finite element models that included the vascular pores and osteocyte lacunae within the tibial metaphysis of Botox-injected (BTX, n = 8) and saline-injected control (CTRL, n = 8) groups. To quantify fluid flow, lacunar and canalicular porosities were modeled as fluid-saturated poroelastic materials, and boundary conditions were applied to simulate physiological loading. This modeling approach allowed a detailed quantification of the fluid flow velocities around osteocytes in a relatively large volume of bone tissue. The analysis demonstrated that interstitial fluid velocity at the vascular pore surfaces was significantly lower in BTX compared to CTRL because of the decreased vascular canal separation. No significant differences in average fluid velocity were observed at the osteocyte lacunae and no correlation was found between the fluid velocity and the lacunar density, which was significantly lower in BTX. Instead, the lacunar fluid velocity was dependent on the osteocyte's specific position in the bone cortex and its proximity to a vascular pore.
Collapse
Affiliation(s)
- Vittorio Gatti
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Michelle J Gelbs
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Rodrigo B Guerra
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Michael B Gerber
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Susannah P Fritton
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| |
Collapse
|
17
|
Digital Twins for Tissue Culture Techniques—Concepts, Expectations, and State of the Art. Processes (Basel) 2021. [DOI: 10.3390/pr9030447] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Techniques to provide in vitro tissue culture have undergone significant changes during the last decades, and current applications involve interactions of cells and organoids, three-dimensional cell co-cultures, and organ/body-on-chip tools. Efficient computer-aided and mathematical model-based methods are required for efficient and knowledge-driven characterization, optimization, and routine manufacturing of tissue culture systems. As an alternative to purely experimental-driven research, the usage of comprehensive mathematical models as a virtual in silico representation of the tissue culture, namely a digital twin, can be advantageous. Digital twins include the mechanistic of the biological system in the form of diverse mathematical models, which describe the interaction between tissue culture techniques and cell growth, metabolism, and the quality of the tissue. In this review, current concepts, expectations, and the state of the art of digital twins for tissue culture concepts will be highlighted. In general, DT’s can be applied along the full process chain and along the product life cycle. Due to the complexity, the focus of this review will be especially on the design, characterization, and operation of the tissue culture techniques.
Collapse
|
18
|
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
|
19
|
Baron C, Nguyen VH, Naili S, Guivier-Curien C. Interaction of ultrasound waves with bone remodelling: a multiscale computational study. Biomech Model Mechanobiol 2020; 19:1755-1764. [DOI: 10.1007/s10237-020-01306-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 02/07/2020] [Indexed: 12/16/2022]
|
20
|
Putra VDL, Song MJ, McBride-Gagyi S, Chang H, Poole K, Whan R, Dean D, Sansalone V, Knothe Tate ML. Mechanomics Approaches to Understand Cell Behavior in Context of Tissue Neogenesis, During Prenatal Development and Postnatal Healing. Front Cell Dev Biol 2020; 7:354. [PMID: 32010686 PMCID: PMC6979483 DOI: 10.3389/fcell.2019.00354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/05/2019] [Indexed: 01/22/2023] Open
Abstract
Mechanomics represents the natural progression of knowledge at the intersection of mechanics and biology with the aim to codify the role of mechanical environment on biological adaptation. Compared to the mapping of the human genome, the challenge of mapping the mechanome remains unsolved. Solving this grand challenge will require both top down and bottom up R&D approaches using experimental and computational tools to visualize and measure adaptation as it occurs. Akin to a mechanical test of a smart material that changes its mechanical properties and local environment under load, stem cells adapt their shape, cytoskeletal architecture, intrinsic mechanical properties, as well as their own niche, through cytoskeletal adaptation as well as up- and down-regulation of structural proteins that modulate their mechanical milieux. Recent advances in live cell imaging allow for unprecedented study and measurements of displacements, shape and volume changes in stem cells, reconfiguring of cytoskeletal machinery (nucleus, cytoskeleton), in response to controlled mechanical forces and stresses applied at cellular boundaries. Coupled with multiphysics computational and virtual power theoretical approaches, these novel experimental approaches enable mechanical testing of stem cells, multicellular templates, and tissues inhabited by stem cells, while the stem cells themselves evolve over time. The novel approach is paving the way to decipher mechanisms of structural and functional adaptation of stem cells in response to controlled mechanical cues. This mini-review outlines integrated approaches and methodologies implemented to date in a series of studies carried out by our consortium. The consortium's body of work is described in context of current roadblocks in the field and innovative, breakthrough solutions and is designed to encourage discourse and cross disciplinary collaboration in the scientific community.
Collapse
Affiliation(s)
- Vina D. L. Putra
- MechBio Team, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Min Jae Song
- MechBio Team, Departments of Biomedical and Mechanical & Aerospace Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
- 3D Bioprinting Core, Ocular and Stem Cell Translational Research Unit, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Sarah McBride-Gagyi
- MechBio Team, Departments of Biomedical and Mechanical & Aerospace Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Hana Chang
- MechBio Team, Departments of Biomedical and Mechanical & Aerospace Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Kate Poole
- Cellular Mechanotransduction Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Renee Whan
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - David Dean
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus, OH, United States
| | - Vittorio Sansalone
- Université Paris-Est Créteil, Laboratoire Modélisation et Simulation Multi Echelle, MSME UMR 8208 CNRS, Créteil Cedex, France
| | - Melissa L. Knothe Tate
- MechBio Team, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- MechBio Team, Departments of Biomedical and Mechanical & Aerospace Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
21
|
Wittig NK, Laugesen M, Birkbak ME, Bach-Gansmo FL, Pacureanu A, Bruns S, Wendelboe MH, Brüel A, Sørensen HO, Thomsen JS, Birkedal H. Canalicular Junctions in the Osteocyte Lacuno-Canalicular Network of Cortical Bone. ACS NANO 2019; 13:6421-6430. [PMID: 31095362 DOI: 10.1021/acsnano.8b08478] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The osteocyte lacuno-canalicular network (LCN) is essential for bone remodeling because osteocytes regulate cell recruitment. This has been proposed to occur through liquid-flow-induced shear forces in the canaliculi. Models of the LCN have thus far assumed that it contains canaliculi connecting the osteocyte lacunae. However, here, we reveal that enlarged spaces occur at places where several canaliculi cross; we name these spaces canalicular junctions. We characterize them in detail within mice cortical bone using synchrotron nanotomography at two length scales, with 50 and 130 nm voxel size, and show that canalicular junctions occur at a density similar to that of osteocyte lacunae and that canalicular junctions tend to cluster. Through confocal laser scanning microscopy, we show that canalicular junctions are widespread as we have observed them in cortical bone from several species, even though the number density of the canalicular junctions was not universal. Fluid flow simulations of a simple model system with and without a canalicular junction clearly show that liquid mass transport and flow velocities are altered by the presence of canalicular junctions. We suggest that these canalicular junctions may play an important role in osteocyte communication and possibly also in canalicular fluid flow. Therefore, we believe that they constitute an important component in the bone osteocyte network.
Collapse
Affiliation(s)
| | | | | | | | | | - Stefan Bruns
- Department of Chemistry, University of Copenhagen , 2100 Copenhagen Ø , Denmark
| | | | | | | | | | | |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The bone is able to adapt its structure to mechanical signals via the bone remodeling process governed by mechanosensitive osteocytes. With aging, an imbalance in bone remodeling results in osteoporosis. In this review, we hypothesized that changes in lacunar morphology underlie the decreased bone mechanoresponsiveness to mechanical loading with aging. RECENT FINDINGS Several studies have reported considerable variations in the shape of osteocytes and their lacunae with aging. Since osteocytes can sense matrix strain directly via their cell bodies, the variations in osteocyte morphology may cause changes in osteocyte mechanosensitivity. As a consequence, the load-adaptive response of osteocytes may change with aging, even when mechanical loading would remain unchanged. Though extensive quantitative data is lacking, evidence exists that the osteocyte lacunae are becoming smaller and more spherical with aging. Future dedicated studies might reveal whether these changes would affect osteocyte mechanosensation and the subsequent biological response, and whether this is (one of) the pathways involved in age-related bone loss.
Collapse
Affiliation(s)
- Haniyeh Hemmatian
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Celestijnenlaan 300c, 3001 Leuven, Belgium
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Astrid D. Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - G. Harry van Lenthe
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Celestijnenlaan 300c, 3001 Leuven, Belgium
| |
Collapse
|
23
|
Ng JL, Kersh ME, Kilbreath S, Knothe Tate M. Establishing the Basis for Mechanobiology-Based Physical Therapy Protocols to Potentiate Cellular Healing and Tissue Regeneration. Front Physiol 2017; 8:303. [PMID: 28634452 PMCID: PMC5460618 DOI: 10.3389/fphys.2017.00303] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Life is mechanobiological: mechanical stimuli play a pivotal role in the formation of structurally and functionally appropriate body templates through mechanobiologically-driven cellular and tissue re/modeling. The body responds to mechanical stimuli engendered through physical movement in an integrated fashion, internalizing and transferring forces from organ, through tissue and cellular length scales. In the context of rehabilitation and therapeutic outcomes, such mechanical stimuli are referred to as mechanotherapy. Physical therapists use mechanotherapy and mechanical interventions, e.g., exercise therapy and manual mobilizations, to restore function and treat disease and/or injury. While the effect of directed movement, such as in physical therapy, is well documented at the length scale of the body and its organs, a number of recent studies implicate its integral effect in modulating cellular behavior and subsequent tissue adaptation. Yet the link between movement biomechanics, physical therapy, and subsequent cellular and tissue mechanoadaptation is not well established in the literature. Here we review mechanoadaptation in the context of physical therapy, from organ to cell scale mechanotransduction and cell to organ scale extracellular matrix genesis and re/modeling. We suggest that physical therapy can be developed to harness the mechanosensitivity of cells and tissues, enabling prescriptive definition of physical and mechanical interventions to enhance tissue genesis, healing, and rehabilitation.
Collapse
Affiliation(s)
- Joanna L. Ng
- Graduate School of Biomedical Engineering, University of New South WalesSydney, NSW, Australia
| | - Mariana E. Kersh
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-ChampaignChampaign, IL, United States
| | - Sharon Kilbreath
- Faculty of Health Sciences, University of SydneySydney, NSW, Australia
| | - M. Knothe Tate
- Graduate School of Biomedical Engineering, University of New South WalesSydney, NSW, Australia
| |
Collapse
|
24
|
Wu X, Wang N, Wang Z, Yu W, Wang Y, Guo Y, Chen W. Mathematically modeling fluid flow and fluid shear stress in the canaliculi of a loaded osteon. Biomed Eng Online 2016; 15:149. [PMID: 28155688 PMCID: PMC5260136 DOI: 10.1186/s12938-016-0267-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Mechanical load-induced intraosseous pressure gradients may result in some fluid stimuli effects, such as fluid flow and fluid shear stress (FSS), which may enable bone cells to detect external mechanical signals. Interstitial bone fluid flow is known to occur in lacunar–canalicular porosity (PLC). Methods In order to characterize lacunar–canalicular fluid flow behavior, a hierarchical osteon system is developed. The osteon is modeled as a poroelastic annular cylinder with two types of impermeable boundary cases considered on its outer wall: one is elastic restrained (Case I), whereas the other is displacement confined (Case II). Analytical solutions such as canalicular fluid velocity, pressure, fluid flow rate (FFR), and shear stress are obtained. Results Results show that the amplitudes of FFR and FSS are proportional to strain amplitude and frequency. However, the key loading factor governing canalicular fluid flow behavior is the strain rate. The larger canalicular radius is, the larger amplitudes of FFR and FSS generalized, especially, the FSS amplitude is proportional to canalicular radius. In addition, both FFR and FSS amplitudes produced in case II are larger than those of case I. Conclusion Strain rate can be acted as a representative loading parameter governing the canalicular fluid flow behavior under a physiological state. This model can facilitate better understanding the load induced the fluid permeation in the PLC. The approach can also be used to analyze the structure of the proteoglycan matrix in the fluid space surrounding the osteocytic process in the canaliculus.
Collapse
Affiliation(s)
- Xiaogang Wu
- Shanxi Key Lab. of Material Strength & Structural Impact and College of Mechanics, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Ningning Wang
- Shanxi Key Lab. of Material Strength & Structural Impact and College of Mechanics, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Zhaowei Wang
- Shanxi Key Lab. of Material Strength & Structural Impact and College of Mechanics, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Weilun Yu
- Shanxi Key Lab. of Material Strength & Structural Impact and College of Mechanics, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Yanqin Wang
- Shanxi Key Lab. of Material Strength & Structural Impact and College of Mechanics, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Yuan Guo
- Shanxi Key Lab. of Material Strength & Structural Impact and College of Mechanics, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China
| | - Weiyi Chen
- Shanxi Key Lab. of Material Strength & Structural Impact and College of Mechanics, Taiyuan University of Technology, Taiyuan, 030024, People's Republic of China.
| |
Collapse
|
25
|
Giorgi M, Verbruggen SW, Lacroix D. In silico bone mechanobiology: modeling a multifaceted biological system. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 8:485-505. [PMID: 27600060 PMCID: PMC5082538 DOI: 10.1002/wsbm.1356] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/27/2016] [Accepted: 07/27/2016] [Indexed: 12/04/2022]
Abstract
Mechanobiology, the study of the influence of mechanical loads on biological processes through signaling to cells, is fundamental to the inherent ability of bone tissue to adapt its structure in response to mechanical stimulation. The immense contribution of computational modeling to the nascent field of bone mechanobiology is indisputable, having aided in the interpretation of experimental findings and identified new avenues of inquiry. Indeed, advances in computational modeling have spurred the development of this field, shedding new light on problems ranging from the mechanical response to loading by individual cells to tissue differentiation during events such as fracture healing. To date, in silico bone mechanobiology has generally taken a reductive approach in attempting to answer discrete biological research questions, with research in the field broadly separated into two streams: (1) mechanoregulation algorithms for predicting mechanobiological changes to bone tissue and (2) models investigating cell mechanobiology. Future models will likely take advantage of advances in computational power and techniques, allowing multiscale and multiphysics modeling to tie the many separate but related biological responses to loading together as part of a larger systems biology approach to shed further light on bone mechanobiology. Finally, although the ever‐increasing complexity of computational mechanobiology models will inevitably move the field toward patient‐specific models in the clinic, the determination of the context in which they can be used safely for clinical purpose will still require an extensive combination of computational and experimental techniques applied to in vitro and in vivo applications. WIREs Syst Biol Med 2016, 8:485–505. doi: 10.1002/wsbm.1356 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Mario Giorgi
- Department of Oncology and Metabolism and INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| | | | - Damien Lacroix
- INSIGNEO Institute for In Silico Medicine, Department of Mechanical Engineering, University of Sheffield, Sheffield, UK.
| |
Collapse
|
26
|
Guyot Y, Smeets B, Odenthal T, Subramani R, Luyten FP, Ramon H, Papantoniou I, Geris L. Immersed Boundary Models for Quantifying Flow-Induced Mechanical Stimuli on Stem Cells Seeded on 3D Scaffolds in Perfusion Bioreactors. PLoS Comput Biol 2016; 12:e1005108. [PMID: 27658116 PMCID: PMC5033382 DOI: 10.1371/journal.pcbi.1005108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 08/16/2016] [Indexed: 12/13/2022] Open
Abstract
Perfusion bioreactors regulate flow conditions in order to provide cells with oxygen, nutrients and flow-associated mechanical stimuli. Locally, these flow conditions can vary depending on the scaffold geometry, cellular confluency and amount of extra cellular matrix deposition. In this study, a novel application of the immersed boundary method was introduced in order to represent a detailed deformable cell attached to a 3D scaffold inside a perfusion bioreactor and exposed to microscopic flow. The immersed boundary model permits the prediction of mechanical effects of the local flow conditions on the cell. Incorporating stiffness values measured with atomic force microscopy and micro-flow boundary conditions obtained from computational fluid dynamics simulations on the entire scaffold, we compared cell deformation, cortical tension, normal and shear pressure between different cell shapes and locations. We observed a large effect of the precise cell location on the local shear stress and we predicted flow-induced cortical tensions in the order of 5 pN/μm, at the lower end of the range reported in literature. The proposed method provides an interesting tool to study perfusion bioreactors processes down to the level of the individual cell's micro-environment, which can further aid in the achievement of robust bioprocess control for regenerative medicine applications.
Collapse
Affiliation(s)
- Yann Guyot
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Biomechanics Research Unit, Université de Liège, Liège, Belgium
| | - Bart Smeets
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Division of Mechatronics, Biostatistics and Sensors (MeBioS), Leuven, Belgium
- Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Tim Odenthal
- Division of Mechatronics, Biostatistics and Sensors (MeBioS), Leuven, Belgium
| | - Ramesh Subramani
- Division of Mechatronics, Biostatistics and Sensors (MeBioS), Leuven, Belgium
| | - Frank P. Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Herman Ramon
- Division of Mechatronics, Biostatistics and Sensors (MeBioS), Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Biomechanics Research Unit, Université de Liège, Liège, Belgium
- Biomechanics Section, KU Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Joukar A, Niroomand-Oscuii H, Ghalichi F. Numerical simulation of osteocyte cell in response to directional mechanical loadings and mechanotransduction analysis: Considering lacunar-canalicular interstitial fluid flow. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2016; 133:133-141. [PMID: 27393805 DOI: 10.1016/j.cmpb.2016.05.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 05/10/2016] [Accepted: 05/31/2016] [Indexed: 06/06/2023]
Abstract
The osteocyte cell is a bone cell that also functions as a bone mechanosensor. In this work, a three-dimensional (3D) fluid-structure interaction (FSI) model of an osteocyte cell under different mechanical loading conditions was used to obtain a better understanding of osteocyte cell behavior under different physiological conditions. In the current study, both fluid and solid parts of osteocyte cell were considered in order to allow for more accurate results. Five different loading conditions have been applied to the osteocyte cell, and consequently the different interstitial fluid flow velocities and shear stresses have been investigated. Furthermore, using a mathematical model, the change in the stimulus function value with shear stress and NO enzyme was revealed. This work suggests that changes in osteocyte morphology and direction of loadings affect cell stimulation. It was found that cell is mostly stimulated and expanded in the direction experiencing the most shear stress. Finally, the amount of cell stimulation was shown quantitatively and there was strong dependency between stimulus function, shear stress, calcium, and NO concentration.
Collapse
Affiliation(s)
- Amin Joukar
- Mechanical Engineering Faculty, Sahand University of Technology, Sahand New Town, Tabriz, Iran
| | - Hanieh Niroomand-Oscuii
- Mechanical Engineering Faculty, Sahand University of Technology, Sahand New Town, Tabriz, Iran.
| | - Farzan Ghalichi
- Mechanical Engineering Faculty, Sahand University of Technology, Sahand New Town, Tabriz, Iran
| |
Collapse
|
28
|
Verbruggen SW, Vaughan TJ, McNamara LM. Mechanisms of osteocyte stimulation in osteoporosis. J Mech Behav Biomed Mater 2016; 62:158-168. [PMID: 27203269 DOI: 10.1016/j.jmbbm.2016.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/28/2016] [Accepted: 05/03/2016] [Indexed: 01/08/2023]
Abstract
Experimental studies have shown that primary osteoporosis caused by oestrogen-deficiency results in localised alterations in bone tissue properties and mineral composition. Additionally, changes to the lacunar-canalicular architecture surrounding the mechanosensitive osteocyte have been observed in animal models of the disease. Recently, it has also been demonstrated that the mechanical stimulation sensed by osteocytes changes significantly during osteoporosis. Specifically, it was shown that osteoporotic bone cells experience higher maximum strains than healthy bone cells after short durations of oestrogen deficiency. However, in long-term oestrogen deficiency there was no significant difference between bone cells in healthy and normal bone. The mechanisms by which these changes arise are unknown. In this study, we test the hypothesis that complex changes in tissue composition and lacunar-canalicular architecture during osteoporosis alter the mechanical stimulation of the osteocyte. The objective of this research is to employ computational methods to investigate the relationship between changes in bone tissue composition and microstructure and the mechanical stimulation of osteocytes during osteoporosis. By simulating physiological loading, it was observed that an initial decrease in tissue stiffness (of 0.425GPa) and mineral content (of 0.66wt% Ca) relative to controls could explain the mechanical stimulation observed at the early stages of oestrogen deficiency (5 weeks post-OVX) during in situ bone cell loading in an oestrogen-deficient rat model of post-menopausal osteoporosis (Verbruggen et al., 2015). Moreover, it was found that a later increase in stiffness (of 1.175GPa) and mineral content (of 1.64wt% Ca) during long-term osteoporosis (34 weeks post-OVX), could explain the mechanical stimuli previously observed at a later time point due to the progression of osteoporosis. Furthermore, changes in canalicular tortuosity arising during osteoporosis were shown to result in increased osteogenic strain stimulation, though to a lesser extent than has been observed experimentally. The findings of this study indicate that changes in the extracellular environment during osteoporosis, arising from altered mineralisation and lacunar-canalicular architecture, lead to altered mechanical stimulation of osteocytes, and provide an enhanced understanding of changes in bone mechanobiology during osteoporosis.
Collapse
Affiliation(s)
- Stefaan W Verbruggen
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Ted J Vaughan
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Laoise M McNamara
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| |
Collapse
|
29
|
Schaffler MB, Cheung WY, Majeska R, Kennedy O. Osteocytes: master orchestrators of bone. Calcif Tissue Int 2014; 94:5-24. [PMID: 24042263 PMCID: PMC3947191 DOI: 10.1007/s00223-013-9790-y] [Citation(s) in RCA: 338] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 08/22/2013] [Indexed: 12/12/2022]
Abstract
Osteocytes comprise the overwhelming majority of cells in bone and are its only true "permanent" resident cell population. In recent years, conceptual and technological advances on many fronts have helped to clarify the role osteocytes play in skeletal metabolism and the mechanisms they use to perform them. The osteocyte is now recognized as a major orchestrator of skeletal activity, capable of sensing and integrating mechanical and chemical signals from their environment to regulate both bone formation and resorption. Recent studies have established that the mechanisms osteocytes use to sense stimuli and regulate effector cells (e.g., osteoblasts and osteoclasts) are directly coupled to the environment they inhabit-entombed within the mineralized matrix of bone and connected to each other in multicellular networks. Communication within these networks is both direct (via cell-cell contacts at gap junctions) and indirect (via paracrine signaling by secreted signals). Moreover, the movement of paracrine signals is dependent on the movement of both solutes and fluid through the space immediately surrounding the osteocytes (i.e., the lacunar-canalicular system). Finally, recent studies have also shown that the regulatory capabilities of osteocytes extend beyond bone to include a role in the endocrine control of systemic phosphate metabolism. This review will discuss how a highly productive combination of experimental and theoretical approaches has managed to unearth these unique features of osteocytes and bring to light novel insights into the regulatory mechanisms operating in bone.
Collapse
Affiliation(s)
- Mitchell B. Schaffler
- University: City College of New York, Department: Biomedical Engineering, Phone: 212-650-5070, Fax: 212-650-6727
| | - Wing-Yee Cheung
- University: City College of New York, Department: Biomedical Engineering
| | - Robert Majeska
- University: City College of New York, Department: Biomedical Engineering
| | - Oran Kennedy
- University: New York University, Department: Orthopaedic Surgery
| |
Collapse
|
30
|
Vaughan TJ, Verbruggen SW, McNamara LM. Are all osteocytes equal? Multiscale modelling of cortical bone to characterise the mechanical stimulation of osteocytes. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2013; 29:1361-1372. [PMID: 23897701 DOI: 10.1002/cnm.2578] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 06/18/2013] [Accepted: 06/20/2013] [Indexed: 06/02/2023]
Abstract
Bone continuously adapts its internal structure to accommodate the functional demands of its mechanical environment. This process is orchestrated by a network of mechanosensitive osteocytes that respond to external mechanical signals and recruit osteoblasts and osteoclasts to alter bone mass to meet loading demands. Because of the irregular hierarchical microarchitecture of bone tissue, the precise mechanical stimuli experienced by osteocytes located in different regions of the tissue is not well-understood. The objective of this study is to characterise the local stimulus experienced by osteocytes distributed throughout the tissue structure. Our models predict that an inhomogeneous microstructural strain field contributes to osteocytes receiving vastly different stimuli at the cellular level, depending on their location within the microstructure. In particular, osteocytes located directly adjacent to micropores experienced strain amplifications in their processes of up to nine times the applied global strain. Furthermore, it was found that the principal orientation of lamellar regions was found to contribute significantly to the magnitude of the stimulus being received at the cellular level. These findings indicate that osteocytes are not equal in terms of the mechanical stimulus being received, and we propose that only a subset of osteocytes may be sufficiently stimulated to function as mechanoreceptors.
Collapse
Affiliation(s)
- Ted J Vaughan
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland; National Centre for Biomedical Engineering Sciences (NCBES), National University of Ireland, Galway, Ireland
| | | | | |
Collapse
|
31
|
Abstract
Osteoporosis is a very common skeletal disorder characterized by reduced bone mass and altered trabecular microarchitecture that leads to bone fragility and fractures. Such disease is due to alterations of the remodeling process that occurs in the basic multicellular units that are transitory cellular complexes including an osteoclastic phase (osteoclast activation and resorption of microscopic portions of bone), a reversion phase (osteoclast replacement by so-called postosteoclastic cells), and an osteoblastic phase (osteoblastic reconstruction of the resorbed bone matrix till the initial volume is regained). Bone remodeling is regulated by a number of systemic and local factors; among the former, besides physical activity and mechanical stresses, a primary role is played by hormones such as parathyroid hormone, vitamin D metabolites, estrogens, calcitonin, and glucocorticoids; among the latter, several growth factors (macrophage colony-stimulating factor, transforming growth factor β, platelet-derived growth factor, fibroblast growth factor 1, bone morphogenetic protein, and insulin-like growth factor 1), as well as the osteoprotegerin-receptor activator of nuclear factor-κ B ligand system and the sclerostin, play a primary function. The remodeling phases can be evaluated by static and dynamic histomorphometry. Their abnormalities may lead to several osteopathies, the most common of which is osteoporosis (above all senile and postmenopausal), a rather elusive disease chiefly due to its slow development. The use of animal models in its study is emphasized.
Collapse
Affiliation(s)
- Ermanno Bonucci
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Paola Ballanti
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
32
|
Jing J, Ren Y, Zong Z, Liu C, Kamiya N, Mishina Y, Liu Y, Zhou X, Feng JQ. BMP receptor 1A determines the cell fate of the postnatal growth plate. Int J Biol Sci 2013; 9:895-906. [PMID: 24163588 PMCID: PMC3807016 DOI: 10.7150/ijbs.7508] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 09/16/2013] [Indexed: 02/05/2023] Open
Abstract
Bone morphogenic proteins (BMPs) are critical for both chondrogenesis and osteogenesis. Previous studies reported that embryos deficient in Bmp receptor (Bmpr)1a or Bmpr1b in cartilage display subtle skeletal defects; however, double mutant embryos develop severe skeletal defects, suggesting a functional redundancy that is essential for early chondrogenesis. In this study, we examined the postnatal role of Bmpr1a in cartilage. In the Bmpr1a conditional knockout (cKO, a cross between Bmpr1a flox and aggrecan-CreERT2 induced by a one-time-tamoxifen injection at birth and harvested at ages of 2, 4, 8 and 20 weeks), there was essentially no long bone growth with little expression of cartilage markers such as SOX9, IHH and glycoproteins. Unexpectedly, the null growth plate was replaced by bone-like tissues, supporting the notions that the progenitor cells in the growth plate, which normally form cartilage, can form other tissues such as bone and fibrous; and that BMPR1A determines the cell fate. A working hypothesis is proposed to explain the vital role of BMPR1A in postnatal chondrogenesis.
Collapse
Affiliation(s)
- Junjun Jing
- 1. State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, China. ; 2. Department of Biomedical Sciences, Texas A&M Baylor College of Dentistry, Dallas, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Piattelli A, Artese L, Penitente E, Iaculli F, Degidi M, Mangano C, Shibli JA, Coelho PG, Perrotti V, Iezzi G. Osteocyte density in the peri-implant bone of implants retrieved after different time periods (4 weeks to 27 years). J Biomed Mater Res B Appl Biomater 2013; 102:239-43. [PMID: 24106071 DOI: 10.1002/jbm.b.33000] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/29/2013] [Accepted: 06/10/2013] [Indexed: 01/22/2023]
Abstract
Bone tissue is characterized by a constant turnover in response to mechanical stimuli, and osteocytes play an essential role in bone mechanical adaptation. However, little to no information has been published regarding osteocyte density as a function of implantation time in vivo. The aim of this retrospective histological study was to evaluate the osteocyte density of the peri-implant bone in implants retrieved because of different reasons in a time period from 4 weeks to 27 years. A total of 18 samples were included in the present study. Specimens were divided into 3 groups depending on the loading history of the implants: loading between 4 weeks and 7 months (group 1); loading between 1 and 5 years (group 2); loading between 14 and 27 years (group 3). All the samples were histologically evaluated and osteocyte density was obtained using the ratio of the number of osteocytes to the bone-area (mm(2) ). The osteocyte density values significantly increased in the Group 2 (1-5 years) compared with Group 1 (4 weeks-7 months), and significantly decreased in the Group 3 (14-27 years) compared to Group 2. No significant differences were detected between Group 1 and Group 3. The decrease in osteocyte density observed in samples that were in vivo for long periods of time under loading is possibly because of the fact that once the bone structure is well aligned and biomechanically competent, a lower number of osteocytes are necessary to keep the tissue homeostasis under loading.
Collapse
Affiliation(s)
- Adriano Piattelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Worton LE, Ausk BJ, Downey LM, Bain SD, Gardiner EM, Srinivasan S, Gross TS, Kwon RY. Systems-based identification of temporal processing pathways during bone cell mechanotransduction. PLoS One 2013; 8:e74205. [PMID: 24040202 PMCID: PMC3770665 DOI: 10.1371/journal.pone.0074205] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/26/2013] [Indexed: 11/25/2022] Open
Abstract
Bone has long been established to be a highly mechanosensitive tissue. When subjected to mechanical loading, bone exhibits profoundly different anabolic responses depending on the temporal pattern in which the stimulus is applied. This phenomenon has been termed temporal processing, and involves complex signal amplification mechanisms that are largely unidentified. In this study, our goal was to characterize transcriptomic perturbations arising from the insertion of intermittent rest periods (a temporal variation with profound effects on bone anabolism) in osteoblastic cells subjected to fluid flow, and assess the utility of these perturbations to identify signaling pathways that are differentially activated by this temporal variation. At the level of the genome, we found that the common and differential alterations in gene expression arising from the two flow conditions were distributionally distinct, with the differential alterations characterized by many small changes in a large number of genes. Using bioinformatics analysis, we identified distinct up- and down-regulation transcriptomic signatures associated with the insertion of rest intervals, and found that the up-regulation signature was significantly associated with MAPK signaling. Confirming the involvement of the MAPK pathway, we found that the insertion of rest intervals significantly elevated flow-induced p-ERK1/2 levels by enabling a second spike in activity that was not observed in response to continuous flow. Collectively, these studies are the first to characterize distinct transcriptomic perturbations in bone cells subjected to continuous and intermittent stimulation, and directly demonstrate the utility of systems-based transcriptomic analysis to identify novel acute signaling pathways underlying temporal processing in bone cells.
Collapse
Affiliation(s)
- Leah E. Worton
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Brandon J. Ausk
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Leah M. Downey
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Steven D. Bain
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Edith M. Gardiner
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sundar Srinivasan
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ted S. Gross
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ronald Y. Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
35
|
Klein-Nulend J, Bakker AD, Bacabac RG, Vatsa A, Weinbaum S. Mechanosensation and transduction in osteocytes. Bone 2013; 54:182-90. [PMID: 23085083 DOI: 10.1016/j.bone.2012.10.013] [Citation(s) in RCA: 317] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 01/08/2023]
Abstract
The human skeleton is a miracle of engineering, combining both toughness and light weight. It does so because bones possess cellular mechanisms wherein external mechanical loads are sensed. These mechanical loads are transformed into biological signals, which ultimately direct bone formation and/or bone resorption. Osteocytes, since they are ubiquitous in the mineralized matrix, are the cells that sense mechanical loads and transduce the mechanical signals into a chemical response. The osteocytes then release signaling molecules, which orchestrate the recruitment and activity of osteoblasts or osteoclasts, resulting in the adaptation of bone mass and structure. In this review, we highlight current insights in bone adaptation to external mechanical loading, with an emphasis on how a mechanical load placed on whole bones is translated and amplified into a mechanical signal that is subsequently sensed by the osteocytes.
Collapse
Affiliation(s)
- Jenneke Klein-Nulend
- Department of Oral Cell Biology, ACTA-VU University Amsterdam, Research Institute MOVE, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
36
|
Song MJ, Dean D, Knothe Tate ML. Mechanical modulation of nascent stem cell lineage commitment in tissue engineering scaffolds. Biomaterials 2013; 34:5766-75. [PMID: 23660249 DOI: 10.1016/j.biomaterials.2013.04.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 04/10/2013] [Indexed: 12/19/2022]
Abstract
Taking inspiration from tissue morphogenesis in utero, this study tests the concept of using tissue engineering scaffolds as delivery devices to modulate emergent structure-function relationships at early stages of tissue genesis. We report on the use of a combined computational fluid dynamics (CFD) modeling, advanced manufacturing methods, and experimental fluid mechanics (micro-piv and strain mapping) for the prospective design of tissue engineering scaffold geometries that deliver spatially resolved mechanical cues to stem cells seeded within. When subjected to a constant magnitude global flow regime, the local scaffold geometry dictates the magnitudes of mechanical stresses and strains experienced by a given cell, and in a spatially resolved fashion, similar to patterning during morphogenesis. In addition, early markers of mesenchymal stem cell lineage commitment relate significantly to the local mechanical environment of the cell. Finally, by plotting the range of stress-strain states for all data corresponding to nascent cell lineage commitment (95% CI), we begin to "map the mechanome", defining stress-strain states most conducive to targeted cell fates. In sum, we provide a library of reference mechanical cues that can be delivered to cells seeded on tissue engineering scaffolds to guide target tissue phenotypes in a temporally and spatially resolved manner. Knowledge of these effects allows for prospective scaffold design optimization using virtual models prior to prototyping and clinical implementation. Finally, this approach enables the development of next generation scaffolds cum delivery devices for genesis of complex tissues with heterogenous properties, e.g., organs, joints or interface tissues such as growth plates.
Collapse
Affiliation(s)
- Min Jae Song
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Cleveland, OH 44106-7207, USA
| | | | | |
Collapse
|
37
|
Fluid flow in the osteocyte mechanical environment: a fluid–structure interaction approach. Biomech Model Mechanobiol 2013; 13:85-97. [DOI: 10.1007/s10237-013-0487-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 03/26/2013] [Indexed: 10/27/2022]
|
38
|
Correia C, Bhumiratana S, Sousa RA, Reis RL, Vunjak-Novakovic G. Sequential application of steady and pulsatile medium perfusion enhanced the formation of engineered bone. Tissue Eng Part A 2013; 19:1244-54. [PMID: 23259605 DOI: 10.1089/ten.tea.2011.0701] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In native bone, cells experience fluctuating shear forces that are induced by pulsatile interstitial flow associated with habitual loading. We hypothesized that the formation of engineered bone can be augmented by replicating such physiologic stimuli to osteogenic cells cultured in porous scaffolds using bioreactors with medium perfusion. To test this hypothesis, we investigated the effect of fluid flow regime on in vitro bone-like tissue development by human adipose stem cells (hASC) cultivated on porous three-dimensional silk fibroin scaffolds. To this end, we varied the sequential relative durations of steady flow (SF) and pulsatile flow (PF) of culture medium applied over a period of 5 weeks, and evaluated their effect on early stages of bone formation. Porous silk fibroin scaffolds (400-600 μm pore size) were seeded with hASC (30×10(6) cells/mL) and cultured in osteogenic medium under four distinct fluid flow regimes: (1) PF for 5 weeks; (2) SF for 1 week, PF for 4 weeks; (3) SF for 2 weeks, PF for 3 weeks; (4) SF for 5 weeks. The PF was applied in 12 h intervals, with the interstitial velocity fluctuating between 400 and 1200 μm/s at a 0.5 Hz frequency for 2 h, followed by 10 h of SF. In all groups, SF was applied at 400 μm/s. The best osteogenic outcomes were achieved for the sequence of 2 weeks of SF and 3 weeks of PF, as evidenced by gene expression (including the PGE2 mechanotransduction marker), construct compositions, histomorphologies, and biomechanical properties. We thus propose that osteogenesis in hASC and the subsequent early stage bone development involve a mechanism, which detects and responds to the level and duration of hydrodynamic shear forces.
Collapse
Affiliation(s)
- Cristina Correia
- Laboratory for Stem Cells and Tissue Engineering, Department of Biomedical Engineering, Columbia University, New York, New York 10032, USA
| | | | | | | | | |
Collapse
|
39
|
Mapping the mechanome of live stem cells using a novel method to measure local strain fields in situ at the fluid-cell interface. PLoS One 2012; 7:e43601. [PMID: 22970134 PMCID: PMC3438189 DOI: 10.1371/journal.pone.0043601] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 07/25/2012] [Indexed: 12/20/2022] Open
Abstract
During mesenchymal condensation, the initial step of skeletogenesis, transduction of minute mechanical forces to the nucleus is associated with up or down-regulation of genes, ultimately resulting in formation of the skeletal template and appropriate cell lineage commitment. The summation of these biophysical cues affects the cell's shape and fate. Here, we predict and measure surface strain, in live stem cells, in response to controlled delivery of stresses, providing a platform to direct short-term structure - function relationships and long-term fate decisions. We measure local strains on stem cell surfaces using fluorescent microbeads coated with Concanavalin A. During delivery of controlled mechanical stresses, 4-Dimensional (x,y,z,t) displacements of the bound beads are measured as surface strains using confocal microscopy and image reconstruction. Similarly, micro-particle image velocimetry (μ-piv) is used to track flow fields with fluorescent microspheres. The measured flow velocity gradient is used to calculate stress imparted by fluid drag at the surface of the cell. We compare strain measured on cell surfaces with those predicted computationally using parametric estimates of the cell's elastic and shear modulus. Finally, cross-correlating stress - strain data to measures of gene transcription marking lineage commitment enables us to create stress - strain - fate maps, for live stem cells in situ. The studies show significant correlations between live stem cell stress - strain relationships and lineage commitment. The method presented here provides a novel means to probe the live stem cell's mechanome, enabling mechanistic studies of the role of mechanics in lineage commitment as it unfolds.
Collapse
|
40
|
Kaiser J, Lemaire T, Naili S, Sansalone V, Komarova S. Do calcium fluxes within cortical bone affect osteocyte mechanosensitivity? J Theor Biol 2012; 303:75-86. [DOI: 10.1016/j.jtbi.2012.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 01/10/2012] [Accepted: 03/01/2012] [Indexed: 01/15/2023]
|
41
|
Abstract
This chapter describes several methods suitable for mechanically stimulating monolayers of bone cells by fluid shear stress (FSS) in vitro. Fluid flow is generated by pumping culture medium through two parallel plates, one of which contains a monolayer of cells. Methods for measuring nitric oxide production by bone cells in response to FSS are also described.
Collapse
Affiliation(s)
- Carmen Huesa
- Department of Developmental Biology, The Roslin Institute, University of Edinburgh, Edinburgh, UK.
| | | |
Collapse
|
42
|
Lemaire T, Lemonnier S, Naili S. On the paradoxical determinations of the lacuno-canalicular permeability of bone. Biomech Model Mechanobiol 2011; 11:933-46. [DOI: 10.1007/s10237-011-0363-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 12/08/2011] [Indexed: 11/28/2022]
|
43
|
Structural differences in the osteocyte network between the calvaria and long bone revealed by three-dimensional fluorescence morphometry, possibly reflecting distinct mechano-adaptations and sensitivities. Biochem Biophys Res Commun 2011; 417:765-70. [PMID: 22198435 DOI: 10.1016/j.bbrc.2011.12.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/08/2011] [Indexed: 02/02/2023]
Abstract
The structural features of osteocytes and their cellular process network are thought to allow for mechanotransduction from the bone tissue to these cells. This study applied three-dimensional fluorescence microscopy to fixed and decalcified bone specimens to quantitatively compare the osteocytes and their networks between mouse parietal bone and tibia that are physiologically enforced by distinct mechanical loads. The subsequent morphometric analysis by the surface rendering of osteocyte cell bodies revealed the tibia to have relatively enriched cytoplasm in the osteocyte cell body in comparison to the parietal bone. Furthermore, quantitative tracing of the cellular processes in silico demonstrated that the numbers of the cellular processes and their bifurcation points per osteocyte in the tibia were significantly higher than those in the parietal bone. Though the total length of the processes per osteocyte in the tibia was two times longer, its total surface area and total volume were smaller than those in the parietal bone, due to its thinner diameter. These architectural differences in the osteocytes and their networks are thus implicated in the adaptation to physiologically different loading, and may also induce distinct mechanosensitivities.
Collapse
|
44
|
Thompson WR, Majid AS, Czymmek KJ, Ruff AL, García J, Duncan RL, Farach-Carson MC. Association of the α(2)δ(1) subunit with Ca(v)3.2 enhances membrane expression and regulates mechanically induced ATP release in MLO-Y4 osteocytes. J Bone Miner Res 2011; 26:2125-39. [PMID: 21638318 PMCID: PMC4478606 DOI: 10.1002/jbmr.437] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Voltage-sensitive calcium channels (VSCCs) mediate signaling events in bone cells in response to mechanical loading. Osteoblasts predominantly express L-type VSCCs composed of the α(1) pore-forming subunit and several auxiliary subunits. Osteocytes, in contrast, express T-type VSCCs and a relatively small amount of L-type α(1) subunits. Auxiliary VSCC subunits have several functions, including modulating gating kinetics, trafficking of the channel, and phosphorylation events. The influence of the α(2)δ auxiliary subunit on T-type VSCCs and the physiologic consequences of that association are incompletely understood and have yet to be investigated in bone. In this study we postulated that the auxiliary α(2) δ subunit of the VSCC complex modulates mechanically regulated ATP release in osteocytes via its association with the T-type Ca(v) 3.2 (α(1H) ) subunit. We demonstrated by reverse-transcriptase polymerase chain reaction, Western blotting, and immunostaining that MLO-Y4 osteocyte-like cells express the T-type Ca(v)3.2(α(1H)) subunit more abundantly than the L-type Ca(v)1.2 (α(1C)) subunit. We also demonstrated that the α(2) δ(1) subunit, previously described as an L-type auxiliary subunit, complexes with the T-type Ca(v)3.2 (α(1H)) subunit in MLO-Y4 cells. Interestingly, siRNA-mediated knockdown of α(2) δ(1) completely abrogated ATP release in response to membrane stretch in MLO-Y4 cells. Additionally, knockdown of the α(2)δ(1) subunit resulted in reduced ERK1/2 activation. Together these data demonstrate a functional VSCC complex. Immunocytochemistry following α(2)δ(1) knockdown showed decreased membrane localization of Ca(v) 3.2 (α(1H)) at the plasma membrane, suggesting that the diminished ATP release and ERK1/2 activation in response to membrane stretch resulted from a lack of Ca(v) 3.2 (α(1H)) at the cell membrane.
Collapse
Affiliation(s)
- William R. Thompson
- University of Delaware, Biomechanics and Movement Science Program, Newark DE, 19716
- University of Delaware, Department of Biological Sciences, Newark DE, 19716
| | - Amber S. Majid
- University of Delaware, Department of Biological Sciences, Newark DE, 19716
| | - Kirk J. Czymmek
- University of Delaware, Department of Biological Sciences, Newark DE, 19716
| | - Albert L. Ruff
- US Army Medical Research Institute of Chemical Defense, Cell and Molecular Biology Branch, Research Division, Aberdeen Proving Ground, MD, 21010
| | - Jesús García
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago IL, 60612
| | - Randall L. Duncan
- University of Delaware, Biomechanics and Movement Science Program, Newark DE, 19716
- University of Delaware, Department of Biological Sciences, Newark DE, 19716
| | - Mary C. Farach-Carson
- University of Delaware, Department of Biological Sciences, Newark DE, 19716
- Rice University, Department of Biochemistry and Cell Biology, Houston, TX, 77005
| |
Collapse
|
45
|
Knothe Tate ML. Top down and bottom up engineering of bone. J Biomech 2010; 44:304-12. [PMID: 21146825 DOI: 10.1016/j.jbiomech.2010.10.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 10/13/2010] [Indexed: 12/15/2022]
Abstract
The goal of this retrospective article is to place the body of my lab's multiscale mechanobiology work in context of top-down and bottom-up engineering of bone. We have used biosystems engineering, computational modeling and novel experimental approaches to understand bone physiology, in health and disease, and across time (in utero, postnatal growth, maturity, aging and death, as well as evolution) and length scales (a single bone like a femur, m; a sample of bone tissue, mm-cm; a cell and its local environment, μm; down to the length scale of the cell's own skeleton, the cytoskeleton, nm). First we introduce the concept of flow in bone and the three calibers of porosity through which fluid flows. Then we describe, in the context of organ-tissue, tissue-cell and cell-molecule length scales, both multiscale computational models and experimental methods to predict flow in bone and to understand the flow of fluid as a means to deliver chemical and mechanical cues in bone. Addressing a number of studies in the context of multiple length and time scales, the importance of appropriate boundary conditions, site specific material parameters, permeability measures and even micro-nanoanatomically correct geometries are discussed in context of model predictions and their value for understanding multiscale mechanobiology of bone. Insights from these multiscale computational modeling and experimental methods are providing us with a means to predict, engineer and manufacture bone tissue in the laboratory and in the human body.
Collapse
Affiliation(s)
- Melissa L Knothe Tate
- Department of Mechanical & Aerospace Engineering, Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106-7222, USA.
| |
Collapse
|
46
|
Schneider P, Meier M, Wepf R, Müller R. Towards quantitative 3D imaging of the osteocyte lacuno-canalicular network. Bone 2010; 47:848-58. [PMID: 20691297 DOI: 10.1016/j.bone.2010.07.026] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 07/26/2010] [Accepted: 07/28/2010] [Indexed: 11/29/2022]
Abstract
Osteocytes are the most abundant cells in bone and the only cells embedded in the bone mineral matrix. They form an extended, three-dimensional (3D) network, whose processes interconnecting the cell bodies reside in thin canals, the canaliculi. Together with the osteocyte lacunae, the canaliculi form the lacuno-canalicular network (LCN). As the negative imprint of the cellular network within bone tissue, the LCN morphology is considered to play a central role for bone mechanosensation and mechanotransduction. However, the LCN has neither been visualized nor quantified in an adequate way up to now. On this account, this article summarizes the current state of knowledge of the LCN morphology and then reviews different imaging methods regarding the quantitative 3D assessment of bone tissue in general and of the LCN in particular. These imaging methods will provide new insights in the field of bone mechanosensation and mechanotransduction and thus, into processes of strain sensation and transduction, which are tightly associated with osteocyte viability and bone quality.
Collapse
|
47
|
Kameo Y, Adachi T, Sato N, Hojo M. Estimation of bone permeability considering the morphology of lacuno-canalicular porosity. J Mech Behav Biomed Mater 2010; 3:240-8. [DOI: 10.1016/j.jmbbm.2009.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 10/08/2009] [Accepted: 10/12/2009] [Indexed: 10/20/2022]
|
48
|
|
49
|
Poroelastic Evaluation of Fluid Movement Through the Lacunocanalicular System. Ann Biomed Eng 2009; 37:1390-402. [DOI: 10.1007/s10439-009-9706-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Accepted: 04/20/2009] [Indexed: 10/20/2022]
|
50
|
Fritton SP, Weinbaum S. Fluid and Solute Transport in Bone: Flow-Induced Mechanotransduction. ANNUAL REVIEW OF FLUID MECHANICS 2009; 41:347-374. [PMID: 20072666 PMCID: PMC2805256 DOI: 10.1146/annurev.fluid.010908.165136] [Citation(s) in RCA: 246] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Much recent evidence suggests that bone cells sense their mechanical environment via interstitial fluid flow. In this review, we summarize theoretical and experimental approaches to quantify fluid and solute transport in bone, starting with the early investigations of fluid shear stress applied to bone cells. The pathways of bone interstitial fluid and solute movement are high-lighted based on recent theoretical models, as well as a new generation of tracer experiments that have clarified and refined the structure and function of the osteocyte pericellular matrix. Then we trace how the fluid-flow models for mechanotransduction have evolved as new ultrastructural features of the osteocyte lacunar-canalicular porosity have been identified and how more recent in vitro fluid-flow and cell-stretch experiments have helped elucidate at the molecular level the possible pathways for cellular excitation in bone.
Collapse
Affiliation(s)
- Susannah P. Fritton
- Department of Biomedical Engineering, City College of New York, New York, New York 10031
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, City College of New York, New York, New York 10031
- Department of Mechanical Engineering, City College of New York, New York, New York 10031
| |
Collapse
|