51
|
Creecy A, Damrath JG, Wallace JM. Control of Bone Matrix Properties by Osteocytes. Front Endocrinol (Lausanne) 2020; 11:578477. [PMID: 33537002 PMCID: PMC7848033 DOI: 10.3389/fendo.2020.578477] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Osteocytes make up 90-95% of the cellular content of bone and form a rich dendritic network with a vastly greater surface area than either osteoblasts or osteoclasts. Osteocytes are well positioned to play a role in bone homeostasis by interacting directly with the matrix; however, the ability for these cells to modify bone matrix remains incompletely understood. With techniques for examining the nano- and microstructure of bone matrix components including hydroxyapatite and type I collagen becoming more widespread, there is great potential to uncover novel roles for the osteocyte in maintaining bone quality. In this review, we begin with an overview of osteocyte biology and the lacunar-canalicular system. Next, we describe recent findings from in vitro models of osteocytes, focusing on the transitions in cellular phenotype as they mature. Finally, we describe historical and current research on matrix alteration by osteocytes in vivo, focusing on the exciting potential for osteocytes to directly form, degrade, and modify the mineral and collagen in their surrounding matrix.
Collapse
Affiliation(s)
- Amy Creecy
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
| | - John G. Damrath
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, United States
- *Correspondence: Joseph M. Wallace,
| |
Collapse
|
52
|
Liu XS, Wang L, de Bakker CMJ, Lai X. Mechanical Regulation of the Maternal Skeleton during Reproduction and Lactation. Curr Osteoporos Rep 2019; 17:375-386. [PMID: 31755029 PMCID: PMC7373497 DOI: 10.1007/s11914-019-00555-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review summarizes recently published data on the effects of pregnancy and lactation on bone structure, mechanical properties, and mechano-responsiveness in an effort to elucidate how the balance between the structural and metabolic functions of the skeleton is achieved during these physiological processes. RECENT FINDINGS While pregnancy and lactation induce significant changes in bone density and structure to provide calcium for fetal/infant growth, the maternal physiology also comprises several innate compensatory mechanisms that allow for the maintenance of skeletal mechanical integrity. Both clinical and animal studies suggest that pregnancy and lactation lead to adaptations in cortical bone structure to allow for rapid calcium release from the trabecular compartment while maintaining whole bone stiffness and strength. Moreover, extents of lactation-induced bone loss and weaning-induced recovery are highly dependent on a given bone's load-bearing function, resulting in better protection of the mechanical integrity at critical load-bearing sites. The recent discovery of lactation-induced osteocytic perilacunar/canalicular remodeling (PLR) indicates a new means for osteocytes to modulate mineral homeostasis and tissue-level mechanical properties of the maternal skeleton. Furthermore, lactation-induced PLR may also play an important role in maintaining the maternal skeleton's load-bearing capacity by altering osteocyte's microenvironment and modulating the transmission of anabolic mechanical signals to osteocytes. Both clinical and animal studies show that parity and lactation have no adverse, or a positive effect on bone strength later in life. The skeletal effects during pregnancy and lactation reflect an optimized balance between the mechanical and metabolic functions of the skeleton.
Collapse
Affiliation(s)
- X Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 332A Stemmler Hall, 36th Street and Hamilton Walk, Philadelphia, PA, USA.
| | - Liyun Wang
- Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Chantal M J de Bakker
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Xiaohan Lai
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
53
|
Does Juvenile Play Programme the Equine Musculoskeletal System? Animals (Basel) 2019; 9:ani9090646. [PMID: 31484397 PMCID: PMC6770595 DOI: 10.3390/ani9090646] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/29/2019] [Accepted: 09/02/2019] [Indexed: 01/21/2023] Open
Abstract
Simple Summary Locomotor play is a common behaviour expressed across a diverse range of species. As a cursorial animal, the horse is capable of locomotor activity within a relatively short time after birth. In the foal, spontaneous locomotor play occurs early in life and has an obvious role in the development of locomotor skills. The intensity and vigour of locomotor play increases with age and this, in turn, provides cumulative increases in the loads the musculoskeletal system experiences. These progressive cumulative loading cycles (bouts of locomotor play), in both the timing and magnitude, reflect the microstrain required to stimulate bone development based on the mechanostat theorem. Data from the published literature were presented to provide empirical support for this hypothesis. Thus, spontaneous locomotor play may be vital to ensure optimal bone development in the horse. Modern production systems need to provide appropriate opportunities for foals to perform spontaneous locomotor play to optimise bone development and reduce the risk of future musculoskeletal injury later in life. Abstract In mammals, play behaviour appears innate and, because of this, may provide insight into the frequency and intensity of load that is required to stimulate positive musculoskeletal development. The objective of this review was to explore the interaction between play and tissue (bone) development at a molecular through to whole-animal level, with specific focus on the horse as a model. The basis of our understanding of the response of bone to loading is the mechanostat theorem. This assumes that at a tissue level, bone attempts to keep localised strain within the physiological range of 1500–2500 microstrain. Loads above this range result in a modelling response to reduce strain, and strain below this threshold results in remodelling to maintain the localised physiological range. In foals, locomotor play is dramatic and vigorous, with cumulative increases in both intensity and complexity. Based on published literature describing locomotor play in foals and the microstrain at different gaits in the horse, it was proposed that locomotor play in foal aligns with the mechanostat theorem in both the magnitude and frequency of load cycles applied. The cumulative increases in the complexity and intensity of locomotor play as the foal develops, in turn, ensure the strain rates associated with play remain above the local physiological range and promote material and architectural changes in the distal limb bones. Thus, spontaneous locomotor play may be vital to ensure optimal bone development in the horse. Modern management systems need to provide appropriate opportunities for foals to perform spontaneous locomotor play to optimise bone development and reduce the risk of future musculoskeletal injury later in life.
Collapse
|
54
|
Abstract
PURPOSE OF REVIEW Osteocytes are the main mechanosensitive cells in bone. Integrin-based adhesions have been shown to facilitate mechanotransduction, and therefore play an important role in load-induced bone formation. This review outlines the role of integrins in osteocyte function (cell adhesion, signalling, and mechanotransduction) and possible role in disease. RECENT FINDINGS Both β1 and β3 integrins subunits have been shown to be required for osteocyte mechanotransduction. Antagonism of these integrin subunits in osteocytes resulted in impaired responses to fluid shear stress. Various disease states (osteoporosis, osteoarthritis, bone metastases) have been shown to result in altered integrin expression and function. Osteocyte integrins are required for normal cell function, with dysregulation of integrins seen in disease. Understanding the mechanism of faulty integrins in disease may aid in the creation of novel therapeutic approaches.
Collapse
Affiliation(s)
- Ivor P Geoghegan
- Department of Mechanical and Biomedical Engineering, Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, National University of Ireland, Galway, Ireland
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - David A Hoey
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2, Ireland
| | - Laoise M McNamara
- Department of Mechanical and Biomedical Engineering, Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, National University of Ireland, Galway, Ireland.
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland.
| |
Collapse
|
55
|
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
|
56
|
Engineered bone for probing organotypic growth and therapy response of prostate cancer tumoroids in vitro. Biomaterials 2019; 197:296-304. [PMID: 30682644 DOI: 10.1016/j.biomaterials.2019.01.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/24/2018] [Accepted: 01/20/2019] [Indexed: 01/23/2023]
Abstract
Mechanistic analysis of metastatic prostate cancer (PCa) biology and therapy response critically depends upon clinically relevant three-dimensional (3D) bone-like, organotypic culture. We here combine an engineered bone-mimetic environment (BME) with longitudinal microscopy to test the growth and therapy response of 3D PCa tumoroids. Besides promoting both tumor-cell autonomous and microenvironment-dependent growth in PCa cell lines and patient-derived xenograft cells, the BME enables in vivo-like tumor cell response to therapy, and reveals bone stroma dependent resistance to chemotherapy and BME-targeted localization and induction of cytoxicity by Radium-223. The BME platform will allow the propagation, compound screening and mechanistic dissection of patient-derived bone tumor isolates and applications toward personalized medicine.
Collapse
|
57
|
Martinez JR, Dhawan A, Farach-Carson MC. Modular Proteoglycan Perlecan/ HSPG2: Mutations, Phenotypes, and Functions. Genes (Basel) 2018; 9:E556. [PMID: 30453502 PMCID: PMC6266596 DOI: 10.3390/genes9110556] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 02/08/2023] Open
Abstract
Heparan sulfate proteoglycan 2 (HSPG2) is an essential, highly conserved gene whose expression influences many developmental processes including the formation of the heart and brain. The gene is widely expressed throughout the musculoskeletal system including cartilage, bone marrow and skeletal muscle. The HSPG2 gene product, perlecan is a multifunctional proteoglycan that preserves the integrity of extracellular matrices, patrols tissue borders, and controls various signaling pathways affecting cellular phenotype. Given HSPG2's expression pattern and its role in so many fundamental processes, it is not surprising that relatively few gene mutations have been identified in viable organisms. Mutations to the perlecan gene are rare, with effects ranging from a relatively mild condition to a more severe and perinatally lethal form. This review will summarize the important studies characterizing mutations and variants of HSPG2 and discuss how these genomic modifications affect expression, function and phenotype. Additionally, this review will describe the clinical findings of reported HSPG2 mutations and their observed phenotypes. Finally, the evolutionary aspects that link gene integrity to function are discussed, including key findings from both in vivo animal studies and in vitro systems. We also hope to facilitate discussion about perlecan/HSPG2 and its role in normal physiology, to explain how mutation can lead to pathology, and to point out how this information can suggest pathways for future mechanistic studies.
Collapse
Affiliation(s)
- Jerahme R Martinez
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Akash Dhawan
- Department of Bioengineering, Rice University, Houston, TX 77005, USA.
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA.
| | - Mary C Farach-Carson
- Department of Bioengineering, Rice University, Houston, TX 77005, USA.
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA.
| |
Collapse
|
58
|
Min S, Lee T, Lee SH, Hong J. Theoretical study of the effect of piezoelectric bone matrix on transient fluid flow in the osteonal lacunocanaliculae. J Orthop Res 2018; 36:2239-2249. [PMID: 29460291 DOI: 10.1002/jor.23873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 01/30/2018] [Indexed: 02/04/2023]
Abstract
A new theoretical generation mechanism of the transient streaming potential considering variations in the surface potential on the wall in a lacunocanalicular system, is proposed based on the assumption of the piezoelectric bone matrix. To obtain the streaming potential analytically, a modified transient charge density equation is proposed. An osteon is modeled as a piezoelectric solid phase having fluid-filled cavities (lacunae) connected by channels (canaliculae) to obtain the pressure gradients in the canaliculae and the electric boundary conditions on the canalicular walls. In addition, this study focused on modeling of the negatively charged glycocalyx that fills the annular fluid space between the osteocytic process and the canalicular wall. It is assumed that the annular fluid space of the canaliculi can be represented as a two-layer configuration for flow through a gap (between the tips of the glycocalyx and the canalicular wall) overlaying the porous glycocalyx. The transient streaming potential and bone fluid flow affected by the generated total potential are analyzed using the one-dimensional lacunocanalicular fluid path, which is surrounded by the piezoelectric bone matrix. A significant increase in the streaming potential is predicted for the case with piezoelectricity. The peak streaming potential value with the piezoelectricity is found to be up to 58.8% greater compared with that without piezoelectricity. The electroviscous effect due to the total electric potential gradients on the fluid velocities in the canaliculi is negligible. These findings imply that the piezoelectric effect caused by deformation of the bone matrix should be considered for prediction of the streaming potential in the lacunocanaliculae. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Sungki Min
- R&D Center, BNR, Seoul, Republic of Korea
| | - Taekyeong Lee
- Department of Control and Instrumentation Engineering, Korea University, Sejong, Republic of Korea
| | - Soon Hyuck Lee
- Department of Orthopedic Surgery, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Junghwa Hong
- Department of Control and Instrumentation Engineering, Korea University, Sejong, Republic of Korea
- Department of Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| |
Collapse
|
59
|
Middleton K, Kondiboyina A, Borrett M, Cui Y, Mei X, You L. Microfluidics approach to investigate the role of dynamic similitude in osteocyte mechanobiology. J Orthop Res 2018; 36:663-671. [PMID: 29027748 DOI: 10.1002/jor.23773] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 10/06/2017] [Indexed: 02/04/2023]
Abstract
Fluid flow is an important regulator of cell function and metabolism in many tissues. Fluid shear stresses have been used to level the mechanical stimuli applied in vitro with what occurs in vivo. However, these experiments often lack dynamic similarity, which is necessary to ensure the validity of the model. For interstitial fluid flow, the major requirement for dynamic similarity is the Reynolds number (Re), the ratio of inertial to viscous forces, is the same between the system and model. To study the necessity of dynamic similarity for cell mechanotransduction studies, we investigated the response of osteocyte-like MLO-Y4 cells to different Re flows at the same level of fluid shear stress. Osteocytes were chosen for this study as flows applied in vitro and in vivo have Re that are orders of magnitude different. We hypothesize that osteocytes' response to fluid flow is Re dependent. We observed that cells exposed to lower and higher Re flows developed rounded and triangular morphologies, respectively. Lower Re flows also reduced apoptosis rates compared to higher Re flows. Furthermore, MLO-Y4 cells exposed to higher Re flows had stronger calcium responses compared to lower Re flows. However, by also controlling for flow rate, the lower Re flows induced a stronger calcium response; while degradation of components of the osteocyte glycocalyx reversed this effect. This work suggests that osteocytes are highly sensitive to differences in Re, independent of just shear stresses, supporting the need for improved in vitro flow platforms that better recapitulate the physiological environment. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:663-671, 2018.
Collapse
Affiliation(s)
- Kevin Middleton
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Room 407, Toronto, Ontario, M5S 3G9, Canada
| | - Avinash Kondiboyina
- Division of Engineering Science, University of Toronto, 40 Saint George Street, Room 2110, Toronto, Ontario, M5S 2E4, Canada
| | - Michael Borrett
- Department of Biochemistry, McMaster University, 1280 Main Street West, Room 4N59, Hamilton, Ontario, L8S 4L8, Canada
| | - Yi Cui
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Room 105, Toronto, Ontario, M5S 3G8, Canada
| | - Xueting Mei
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Room 105, Toronto, Ontario, M5S 3G8, Canada
| | - Lidan You
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Room 407, Toronto, Ontario, M5S 3G9, Canada.,Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Room 105, Toronto, Ontario, M5S 3G8, Canada
| |
Collapse
|
60
|
Wang B, Sun X, Akkus O, Wang L. Elevated solute transport at sites of diffuse matrix damage in cortical bone: Implications on bone repair. J Orthop Res 2018; 36:692-698. [PMID: 28921632 PMCID: PMC5839948 DOI: 10.1002/jor.23742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/13/2017] [Indexed: 02/04/2023]
Abstract
UNLABELLED Diffuse matrix damage in rat cortical bone has been observed to self-repair efficiently in 2 weeks without activating bone remodeling, and unlike the case with linear cracks, the local osteocytes at the sites of diffuse damage remain healthy. However, the reason(s) for such high efficiency of matrix repair remains unclear. We hypothesized that transport of minerals and other compounds essential for damage repair is enhanced at the damaged sites and further increased by the application of tensile loading. To test our hypothesis, diffuse damage was introduced in notched bovine wafers under cyclic tensile loading and unloading. Using the Fluorescence Recovery After Photobleaching (FRAP) approach, we measured the transport of a small fluorescent tracer (sodium fluorescein, 376 Da) in damaged versus undamaged regions and under varying tensile load magnitudes (0.2 N, 10 N, 20 N, and 30 N), which corresponded to nominal strains of 12.5, 625, 1,250, and 1,875 microstrains, respectively. We found a 37% increase in transport of fluorescein in damaged regions relative to undamaged regions and a further ∼18% increase in transport under 20 N and 30 N tension compared to the non-loaded condition, possibly due to the opening of the cracking surfaces. The elevated transport of minerals and other adhesive proteins may, at least partially, account for the highly effective repair of diffuse damage observed in vivo. CLINICAL SIGNIFICANCE Diffuse damage adversely affects bone's fracture resistance and this study provided quantitative data on elevated transport, which may be involved in repairing diffuse damage in vivo. 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:692-698, 2018.
Collapse
Affiliation(s)
- Bin Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA,Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, P.R. China,Corresponding Authors: Liyun Wang, Ph.D., 130 Academy Street, University of Delaware, Newark, DE 19716, Phone: (302)-831-2659, Fax: (302)-831-3619, ; Bin Wang, Ph.D., No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China, Phone: 86-23-63662443, Fax: 86-23-68485111,
| | - Xuanhao Sun
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Ozan Akkus
- Department of Mechanical and Aerospace Engineering, and Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA,Corresponding Authors: Liyun Wang, Ph.D., 130 Academy Street, University of Delaware, Newark, DE 19716, Phone: (302)-831-2659, Fax: (302)-831-3619, ; Bin Wang, Ph.D., No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China, Phone: 86-23-63662443, Fax: 86-23-68485111,
| |
Collapse
|
61
|
Abstract
PURPOSE OF REVIEW Solute transport in the lacunar-canalicular system (LCS) plays important roles in osteocyte metabolism and cell-cell signaling. This review will summarize recent studies that establish pericellular matrix (PCM), discovered inside the LCS, as a crucial regulator of solute transport in bone. RECENT FINDINGS Utilizing confocal imaging and mathematical modeling, recent studies successfully quantified molecular diffusion and convection in the LCS as well as the size-dependent sieving effects of the PCM, leading to the quantification of the effective PCM fiber spacing (10 to 17 nm) in murine adult bones. Perlecan/HSPG2, a large linear proteoglycan, was identified to be an essential PCM component. The PCM-filled LCS is bone's chromatographic column, where fluid/solute transport to and from the osteocytes is regulated. The chemical composition, deposition rate, and turnover rate of the osteocyte PCM should be further defined to better understand osteocyte physiology and bone metabolism.
Collapse
Affiliation(s)
- Liyun Wang
- Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, 130 Academy Street, Newark, DE, 19716, USA.
| |
Collapse
|
62
|
Yu K, Sellman DP, Bahraini A, Hagan ML, Elsherbini A, Vanpelt KT, Marshall PL, Hamrick MW, McNeil A, McNeil PL, McGee-Lawrence ME. Mechanical loading disrupts osteocyte plasma membranes which initiates mechanosensation events in bone. J Orthop Res 2018; 36:653-662. [PMID: 28755471 PMCID: PMC5788741 DOI: 10.1002/jor.23665] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/24/2017] [Indexed: 02/04/2023]
Abstract
Osteocytes sense loading in bone, but their mechanosensation mechanisms remain poorly understood. Plasma membrane disruptions (PMD) develop with loading under physiological conditions in many cell types (e.g., myocytes, endothelial cells). These PMD foster molecular flux across cell membranes that promotes tissue adaptation, but this mechanosensation mechanism had not been explored in osteocytes. Our goal was to investigate whether PMD occur and initiate consequent mechanotransduction in osteocytes during physiological loading. We found that osteocytes experience PMD during in vitro (fluid flow) and in vivo (treadmill exercise) mechanical loading, in proportion to the level of stress experienced. In fluid flow studies, osteocyte PMD preferentially formed with rapid as compared to gradual application of loading. In treadmill studies, osteocyte PMD increased with loading in weight bearing locations (tibia), but this trend was not seen in non-weight bearing locations (skull). PMD initiated osteocyte mechanotransduction including calcium signaling and expression of c-fos, and repair rates of these PMD could be enhanced or inhibited pharmacologically to alter downstream mechanotransduction and osteocyte survival. PMD may represent a novel mechanosensation pathway in bone and a target for modifying skeletal adaptation signaling in osteocytes. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:653-662, 2018.
Collapse
Affiliation(s)
- Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - David P. Sellman
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Anoosh Bahraini
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Mackenzie L. Hagan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Ahmed Elsherbini
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Kayce T. Vanpelt
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Peyton L. Marshall
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Mark W. Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Anna McNeil
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Paul L. McNeil
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| | - Meghan E. McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, 1120 15 St, Augusta, GA
| |
Collapse
|
63
|
Microstructural changes associated with osteoporosis negatively affect loading-induced fluid flow around osteocytes in cortical bone. J Biomech 2017; 66:127-136. [PMID: 29217091 DOI: 10.1016/j.jbiomech.2017.11.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 11/05/2017] [Accepted: 11/09/2017] [Indexed: 12/21/2022]
Abstract
Loading-induced interstitial fluid flow in the microporosities of bone is critical for osteocyte mechanotransduction and for the maintenance of tissue health, enhancing convective transport in the lacunar-canalicular system. In recent studies, our group has reported alterations of bone's vascular porosity and lacunar-canalicular system microarchitecture in a rat model of postmenopausal osteoporosis. In this work, poroelastic finite element analysis was used to investigate whether these microstructural changes can affect interstitial fluid flow around osteocytes. Animal-specific finite element models were developed combining micro-CT reconstructions of bone microstructure and measures of the poroelastic material properties. These models were used to quantify and compare loading-induced fluid flow in the lacunar-canalicular system of ovariectomized and sham-operated rats. A parametric analysis was also used to quantify the influence of the lacunar-canalicular permeability and vascular porosity on the fluid velocity magnitude. Results show that mechanically-induced interstitial fluid velocity can be significantly reduced in the lacunar-canalicular system of ovariectomized rats. Interestingly, the vascular porosity is shown to have a major influence on interstitial fluid flow, while the lacunar-canalicular permeability influence is limited when larger than 10-20m2. Altogether our results suggest that microstructural changes associated with the osteoporotic condition can negatively affect interstitial fluid flow around osteocytes in the lacunar-canalicular system of cortical bone. This fluid flow reduction could impair mechanosensation of the osteocytic network, possibly playing a role in the initiation and progression of age-related bone loss and postmenopausal osteoporosis.
Collapse
|
64
|
Zhang B, Hou R, Zou Z, Luo T, Zhang Y, Wang L, Wang B. Mechanically induced autophagy is associated with ATP metabolism and cellular viability in osteocytes in vitro. Redox Biol 2017; 14:492-498. [PMID: 29096322 PMCID: PMC5680519 DOI: 10.1016/j.redox.2017.10.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/17/2017] [Accepted: 10/25/2017] [Indexed: 12/16/2022] Open
Abstract
Both mechanical loading and intracellular autophagy play important roles in bone homeostasis; however, their relationship remains largely unexplored. The objectives of this study were to determine whether osteocytes undergo autophagy upon fluid shear stress (FSS) loading and to determine the correlation between mechanically induced autophagy and ATP metabolism. Autophagic vacuoles were observed by transmission electron microscopy (TEM) in osteocyte-like MLO-Y4 cells subjected to FSS. Increased autophagic flux was further confirmed by the increased amount of the LC3-II isoform and the degradation of p62. Fluorescent puncta distributed in the cytoplasm were observed in the GFP-LC3 transformed cells subjected to FSS. Furthermore, FSS-induced ATP release and synthesis in osteocytes were attenuated by inhibiting autophagy with 3-MA. After FSS exposure, a high ratio of cell death was observed in cultures pretreated with 3-MA, an autophagy inhibitor, with no significantly different Caspase 3/7 activity. Our results indicated that FSS induces protective autophagy in osteocytes and that mechanically induced autophagy is associated with ATP metabolism and osteocyte survival. From the clinical perspective, it may be possible to enhance skeletal cell survival with drugs that modulate the autophagic state, and the autophagy-related pathway could be a potential target for the prevention of ageing-related bone disorders. Fluid flow shear stress (FSS) induces activation of autophagic flux in MLO-Y4 osteocytes. FSS-induced autophagy promoted ATP metabolism in MLO-Y4 osteocytes. Inhibited autophagy decreased FSS-induced ATP release. FSS-induced autophagy was beneficial to the osteocyte survival after FSS exposure.
Collapse
Affiliation(s)
- Bingbing Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Rutao Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zhen Zou
- Department of Medical Laboratory Technology, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Tiantian Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yang Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Bin Wang
- Department of Medical Laboratory Technology, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
65
|
Bertacchini J, Benincasa M, Checchi M, Cavani F, Smargiassi A, Ferretti M, Palumbo C. Expression and functional proteomic analyses of osteocytes from Xenopus laevis tested under mechanical stress conditions: preliminary observations on an appropriate new animal model. J Anat 2017; 231:823-834. [PMID: 28925539 DOI: 10.1111/joa.12685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2017] [Indexed: 12/12/2022] Open
Abstract
Hitherto, the role of the osteocyte as transducer of mechanical stimuli into biological signals is far from settled. In this study, we used an appropriate model represented by the cortex of Xenopus laevis long bone diaphysis lacking (unlike the mammalian one) of vascular structures and containing only osteocytes inside the bone matrix. These structural features allow any change of protein profile that might be observed upon different experimental conditions, such as bone adaptation to stress/mechanical loading, to be ascribed specifically to osteocytes. The study was conducted by combining ultrastructural observations and two-dimensional electrophoresis for proteomic analysis. The osteocyte population was extracted from long bones of lower limbs of amphibian skeletons after different protocols (free and forced swimming). The experiments were performed on 210 frogs subdivided into five trials, each including free swimming frogs (controls) and frogs submitted to forced swimming (stressed). The stressed groups were obliged to swim (on movable spheres covering the bottom of a pool on a vibrating plate) continuously for 8 h, and killed 24 h later along with the control groups. Long bones free of soft tissues (periosteum, endosteum and bone marrow), as well as muscles of posterior limbs, were processed and analyzed for proteins differentially expressed or phosphorylated between the two sample groups. The comparative analysis showed that protein phosphorylation profiles differ between control and stressed groups. In particular, we found in long bones of stressed samples that both Erk1/2 and Akt are hyperphosphorylated; moreover, the different phosphorylation of putative Akt substrates (recognized by specific Akt phosphosubstrates-antibody) in stressed vs. control samples clearly demonstrated that Akt signaling is boosted by forced swimming (leading to an increase of mechanical stress) of amphibian long bones. In parallel, we found in posterior limb muscles that the expression of heat shock protein HSP27 and HSP70 stress markers increased upon the forced swimming condition. Because the cortexes of frog long bones are characterized by the presence of only osteocytes, all our results establish the suitability of the X. laevis animal model to study the bone response to stress conditions mediated by this cell type and pave the way for further analysis of the signaling pathways involved in these signal transduction mechanisms.
Collapse
Affiliation(s)
- Jessika Bertacchini
- Dipartimento di Scienze Biomediche Metaboliche e Neuroscienze, Sezione di Morfologia umana. Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Marta Benincasa
- Dipartimento di Scienze Biomediche Metaboliche e Neuroscienze, Sezione di Morfologia umana. Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Marta Checchi
- Dipartimento di Scienze Biomediche Metaboliche e Neuroscienze, Sezione di Morfologia umana. Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Francesco Cavani
- Dipartimento di Scienze Biomediche Metaboliche e Neuroscienze, Sezione di Morfologia umana. Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Alberto Smargiassi
- Dipartimento di Scienze Biomediche Metaboliche e Neuroscienze, Sezione di Morfologia umana. Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Marzia Ferretti
- Dipartimento di Scienze Biomediche Metaboliche e Neuroscienze, Sezione di Morfologia umana. Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Carla Palumbo
- Dipartimento di Scienze Biomediche Metaboliche e Neuroscienze, Sezione di Morfologia umana. Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| |
Collapse
|
66
|
Xu Z, Chen J, Shao W, Wang R, Liu Y. [Research progress in osteogenesis and osteogenic mechanism of heparan sulfate]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:1016-1020. [PMID: 29806444 PMCID: PMC8458588 DOI: 10.7507/1002-1892.201701103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/13/2017] [Indexed: 11/03/2022]
Abstract
Objective To discuss the role of heparan sulfate (HS) in bone formation and bone remodeling and summarize the research progress in the osteogenic mechanism of HS. Methods The domestic and abroad related literature about HS acting on osteoblast cell line in vitro, HS and HS composite scaffold materials acting on the ani-mal bone defect models, and the effect of HS proteoglycans on bone development were summarized and analyzed. Results Many growth factors involved in fracture healing especially heparin-binding growth factors, such as fibroblast growth factors, bone morphogenetic protein, and transforming growth factor β, are connected noncovalently with long HS chains. HS proteoglycans protect these proteins from protease degradation and are directly involved in the regulation of growth factors signaling and bone cell function. HS can promote the differentiation of stem cells into osteoblasts and enhance the differentiation of osteoblasts. In bone matrix, HS plays a significant role in promoting the formation, maintaining the stability, and accelerating the mineralization. Conclusion The osteogenesis of HS is pronounced. HS is likely to become the clinical treatment measures of fracture nonunion or delayed union, and is expected to provide more choices for bone tissue engineering with identification of its long-term safety.
Collapse
Affiliation(s)
- Zhujie Xu
- Department of Orthopedics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi Jiangsu, 214000, P.R.China
| | - Jinghua Chen
- Medicinal Biopolymer Laboratory of College of Pharmacy, Jiangnan University, Wuxi Jiangsu, 214000, P.R.China
| | - Wei Shao
- Department of Orthopedics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi Jiangsu, 214000, P.R.China
| | - Rui Wang
- Department of Orthopedics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi Jiangsu, 214000, P.R.China
| | - Yi Liu
- Department of Orthopedics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi Jiangsu, 214000, P.R.China;Medicinal Biopolymer Laboratory of College of Pharmacy, Jiangnan University, Wuxi Jiangsu, 214000,
| |
Collapse
|
67
|
Papy-Garcia D, Albanese P. Heparan sulfate proteoglycans as key regulators of the mesenchymal niche of hematopoietic stem cells. Glycoconj J 2017; 34:377-391. [PMID: 28577070 DOI: 10.1007/s10719-017-9773-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 12/21/2022]
Abstract
The complex microenvironment that surrounds hematopoietic stem cells (HSCs) in the bone marrow niche involves different coordinated signaling pathways. The stem cells establish permanent interactions with distinct cell types such as mesenchymal stromal cells, osteoblasts, osteoclasts or endothelial cells and with secreted regulators such as growth factors, cytokines, chemokines and their receptors. These interactions are mediated through adhesion to extracellular matrix compounds also. All these signaling pathways are important for stem cell fates such as self-renewal, proliferation or differentiation, homing and mobilization, as well as for remodeling of the niche. Among these complex molecular cues, this review focuses on heparan sulfate (HS) structures and functions and on the role of enzymes involved in their biosynthesis and turnover. HS associated to core protein, constitute the superfamily of heparan sulfate proteoglycans (HSPGs) present on the cell surface and in the extracellular matrix of all tissues. The key regulatory effects of major medullar HSPGs are described, focusing on their roles in the interactions between hematopoietic stem cells and their endosteal niche, and on their ability to interact with Heparin Binding Proteins (HBPs). Finally, according to the relevance of HS moieties effects on this complex medullar niche, we describe recent data that identify HS mimetics or sulfated HS signatures as new glycanic tools and targets, respectively, for hematopoietic and mesenchymal stem cell based therapeutic applications.
Collapse
Affiliation(s)
- Dulce Papy-Garcia
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France
| | - Patricia Albanese
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France.
| |
Collapse
|
68
|
Pacheco-Costa R, Kadakia JR, Atkinson EG, Wallace JM, Plotkin LI, Reginato RD. Connexin37 deficiency alters organic bone matrix, cortical bone geometry, and increases Wnt/β-catenin signaling. Bone 2017; 97:105-113. [PMID: 28096061 DOI: 10.1016/j.bone.2017.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 01/12/2017] [Accepted: 01/12/2017] [Indexed: 10/20/2022]
Abstract
Deletion of connexin (Cx) 37 in mice leads to increased cancellous bone mass due to defective osteoclast differentiation. Paradoxically; however, Cx37-deficient mice exhibit reduced cortical thickness accompanied by higher bone strength, suggesting a contribution of Cx37 to bone matrix composition. Thus, we investigated whether global deletion of Cx37 alters the composition of organic bone extracellular matrix. Five-month-old Cx37-/- mice exhibited increased marrow cavity area, and periosteal and endocortical bone surface resulting in higher total area in tibia compared to Cx37+/+ control mice. Deletion of Cx37 increased genes involved in collagen maturation (loxl3 and loxl4) and glycosaminoglycans- (chsy1, chpf and has3) proteoglycans- associated genes (biglycan and decorin). In addition, expression of type II collagen assessed by immunostaining was increased by 82% whereas collagen maturity by picrosirius-polarizarion tended to be reduced (p=0.071). Expression of glycosaminoglycans by histochemistry was decreased, whereas immunostaining revealed that biglycan was unchanged and decorin was slightly increased in Cx37-/- bone sections. Consistent with these in vivo findings, MLO-Y4 osteocytic cells silenced for Cx37 gene exhibited increased mRNA levels for collagen synthesis (col1a1 and col3a1) and collagen maturation (lox, loxl1 and loxl2 genes). Furthermore, mechanistic studies showed Wnt/β-catenin activation in MLO-Y4 osteocytic cells, L5 vertebra, and authentic calvaria-derived osteocytes isolated by fluorescent-activated cell sorter. Our findings demonstrate that altered profile of the bone matrix components in Cx37-deficient mice acts in favor of higher resistance to fracture in long bones.
Collapse
Affiliation(s)
- Rafael Pacheco-Costa
- Departamento de Morfologia e Genética, Universidade Federal de Sao Paulo - Escola Paulista de Medicina, São Paulo, Brazil; Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Jay R Kadakia
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, IN, USA.
| | - Emily G Atkinson
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, IN, USA; Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA.
| | - Lilian I Plotkin
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
| | - Rejane D Reginato
- Departamento de Morfologia e Genética, Universidade Federal de Sao Paulo - Escola Paulista de Medicina, São Paulo, Brazil.
| |
Collapse
|
69
|
Yang H, Embry RE, Main RP. Effects of Loading Duration and Short Rest Insertion on Cancellous and Cortical Bone Adaptation in the Mouse Tibia. PLoS One 2017; 12:e0169519. [PMID: 28076363 PMCID: PMC5226737 DOI: 10.1371/journal.pone.0169519] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 12/19/2016] [Indexed: 11/19/2022] Open
Abstract
The skeleton's osteogenic response to mechanical loading can be affected by loading duration and rest insertion during a series of loading events. Prior animal loading studies have shown that the cortical bone response saturates quickly and short rest insertions between load cycles can enhance cortical bone formation. However, it remains unknown how loading duration and short rest insertion affect load-induced osteogenesis in the mouse tibial compressive loading model, and particularly in cancellous bone. To address this issue, we applied cyclic loading (-9 N peak load; 4 Hz) to the tibiae of three groups of 16 week-old female C57BL/6 mice for two weeks, with a different number of continuous load cycles applied daily to each group (36, 216 and 1200). A fourth group was loaded under 216 daily load cycles with a 10 s rest insertion after every fourth cycle. We found that as few as 36 load cycles per day were able to induce osteogenic responses in both cancellous and cortical bone. Furthermore, while cortical bone area and thickness continued to increase through 1200 cycles, the incremental increase in the osteogenic response decreased as load number increased, indicating a reduced benefit of the increasing number of load cycles. In the proximal metaphyseal cancellous bone, trabecular thickness increased with load up to 216 cycles. We also found that insertion of a 10 s rest between load cycles did not improve the osteogenic response of the cortical or cancellous tissues compared to continuous loading in this model given the age and sex of the mice and the loading parameters used here. These results suggest that relatively few load cycles (e.g. 36) are sufficient to induce osteogenic responses in both cortical and cancellous bone in the mouse tibial loading model. Mechanistic studies using the mouse tibial loading model to examine bone formation and skeletal mechanobiology could be accomplished with relatively few load cycles.
Collapse
Affiliation(s)
- Haisheng Yang
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Rachel E. Embry
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Russell P. Main
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
70
|
Abstract
Bone, despite its relatively inert appearance, is a tissue that is capable of adapting to its environment. Wolff’s law, first described in the 19th century, describes the ability of bone to change structure depending on the mechanical forces applied to it. The mechanostat model extended this principle and suggested that the amount of strain a bone detects depends on bone strength and the amount of muscle force applied to the bone. Experimental studies have found that low-magnitude, high-frequency mechanical loading is considered to be the most effective at increasing bone formation. The osteocyte is considered to be the master regulator of the bone response to mechanical loading. Deformation of bone matrix by mechanical loading is thought to result in interstitial fluid flow within the lacunar–canalicular system, which may activate osteocyte mechanosensors, leading to changes in osteocyte gene expression and ultimately increased bone formation and decreased bone resorption. However, repetitive strain applied to bone can result in microcracks, which may propagate and coalesce, and if not repaired predispose to catastrophic fracture. Osteocytes are a key component in this process, whereby apoptotic osteocytes in an area of microdamage promote targeted remodeling of the damaged bone. If fractures do occur, fracture repair can be divided into 2 types: primary and secondary healing. Secondary fracture repair is the most common and is a multistage process consisting of hematoma formation and acute inflammation, callus formation, and finally remodeling, whereby bone may return to its original form.
Collapse
|
71
|
Fan L, Pei S, Lucas Lu X, Wang L. A multiscale 3D finite element analysis of fluid/solute transport in mechanically loaded bone. Bone Res 2016; 4:16032. [PMID: 27722020 PMCID: PMC5037578 DOI: 10.1038/boneres.2016.32] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/15/2016] [Accepted: 08/21/2016] [Indexed: 12/02/2022] Open
Abstract
The transport of fluid, nutrients, and signaling molecules in the bone lacunar–canalicular system (LCS) is critical for osteocyte survival and function. We have applied the fluorescence recovery after photobleaching (FRAP) approach to quantify load-induced fluid and solute transport in the LCS in situ, but the measurements were limited to cortical regions 30–50 μm underneath the periosteum due to the constrains of laser penetration. With this work, we aimed to expand our understanding of load-induced fluid and solute transport in both trabecular and cortical bone using a multiscaled image-based finite element analysis (FEA) approach. An intact murine tibia was first re-constructed from microCT images into a three-dimensional (3D) linear elastic FEA model, and the matrix deformations at various locations were calculated under axial loading. A segment of the above 3D model was then imported to the biphasic poroelasticity analysis platform (FEBio) to predict load-induced fluid pressure fields, and interstitial solute/fluid flows through LCS in both cortical and trabecular regions. Further, secondary flow effects such as the shear stress and/or drag force acting on osteocytes, the presumed mechano-sensors in bone, were derived using the previously developed ultrastructural model of Brinkman flow in the canaliculi. The material properties assumed in the FEA models were validated against previously obtained strain and FRAP transport data measured on the cortical cortex. Our results demonstrated the feasibility of this computational approach in estimating the fluid flux in the LCS and the cellular stimulation forces (shear and drag forces) for osteocytes in any cortical and trabecular bone locations, allowing further studies of how the activation of osteocytes correlates with in vivo functional bone formation. The study provides a promising platform to reveal potential cellular mechanisms underlying the anabolic power of exercises and physical activities in treating patients with skeletal deficiencies.
Collapse
Affiliation(s)
- Lixia Fan
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA; School of Mechanical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Shaopeng Pei
- Department of Mechanical Engineering, University of Delaware , Newark, DE, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware , Newark, DE, USA
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware , Newark, DE, USA
| |
Collapse
|
72
|
Barritault D, Desgranges P, Meddahi-Pellé A, Denoix JM, Saffar JL. RGTA ®-based matrix therapy - A new branch of regenerative medicine in locomotion. Joint Bone Spine 2016; 84:283-292. [PMID: 27663756 DOI: 10.1016/j.jbspin.2016.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/10/2016] [Indexed: 10/21/2022]
Abstract
Matrix therapy is an innovative, minimally invasive approach in the field of regenerative medicine, that aims to promote tissue regeneration by reconstructing the cellular microenvironment following tissue injury. This approach has significant therapeutic potential in the treatment of pathologies characterized by tissue inflammation and damage, or following injury, conditions which can be incapacitating and cost-consuming. Heparan sulfate mimics, termed ReGeneraTing Agents (RGTA®s) have emerged as a unifying approach to treat these diverse pathologies. Today, skin and corneal healing topical products have already been used in clinics, demonstrating a proof of concept in humans. In this review, we present key evidence that RGTA®s regenerate damaged tissue in bone, muscle, tendon and nerve, with astonishing results. In animal models of bone surgical defects and inflammatory bone loss, RGTA® induced healing of injured bones by controlling inflammation and bone resorption, and stimulated bone formation by coordinating vascularization, recruitment and differentiation of competent cells from specific niches, restoring tissue quality to that of uninjured tissue, evoking true regeneration. In models of muscle injury, RGTA® had marked effects on healing speed and quality, evidenced by increased muscle fiber density, maturation, vascularization and reduced fibrosis, more mature motor endplates and functional recovery. Applications merging RGTA®-based matrix therapy and cell therapy, combining Extra-Cellular Matrix reconstruction with cells required for optimal tissue repair show significant promise. Hence restoration of the proper microenvironment is a new paradigm in regenerative medicine. Harnessing the potential of RGTA® in this brave, new vision of regenerative therapy will therefore be the focus of future studies.
Collapse
Affiliation(s)
- Denis Barritault
- Université Paris-Est Créteil, Laboratoire CRRET and OTR3, 4 rue Française, 75001 Paris, 1.2, France.
| | - Pascal Desgranges
- Université Paris-Est-Créteil, Hôpital Henri Mondor, Paris XII, Vascular Surgery Unit, 51, av du MI de Lattre de Tassigny, 94010 Creteil, France
| | - Anne Meddahi-Pellé
- Inserm U1148, LVTS, Université Paris 7, Université Paris 13, Sorbonne Paris Cité, Hôpital Bichat, 46 rue H Huchard, 75018 Paris, France
| | - Jean-Marie Denoix
- Université Paris Est, Ecole Nationale Vétérinaire d'Alfort, USC 957 BPLC, 94700 Maisons-Alfort, France; Centre d'Imagerie et de Recherche sur les Affections Locomotrices Equine (CIRALE), 14430 Goustranville, France
| | - Jean-Louis Saffar
- EA2496 Laboratoire Pathologies, Imagerie et Biothérapies Oro-Faciales, Faculté de Chirurgie Dentaire, Université Paris Descartes, Sorbonne Paris Cité, 1 rue Maurice Arnoux, 92120 Montrouge, France
| |
Collapse
|
73
|
Gubbiotti MA, Neill T, Iozzo RV. A current view of perlecan in physiology and pathology: A mosaic of functions. Matrix Biol 2016; 57-58:285-298. [PMID: 27613501 DOI: 10.1016/j.matbio.2016.09.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/01/2016] [Indexed: 01/06/2023]
Abstract
Perlecan, a large basement membrane heparan sulfate proteoglycan, is expressed in a wide array of tissues where it regulates diverse cellular processes including bone formation, inflammation, cardiac development, and angiogenesis. Here we provide a contemporary review germane to the biology of perlecan encompassing its genetic regulation as well as an analysis of its modular protein structure as it pertains to function. As perlecan signaling from the extracellular matrix converges on master regulators of autophagy, including AMPK and mTOR, via a specific interaction with vascular endothelial growth factor receptor 2, we specifically focus on the mechanism of action of perlecan in autophagy and angiogenesis and contrast the role of endorepellin, the C-terminal fragment of perlecan, in these cellular and morphogenic events.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Thomas Neill
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
74
|
Laurent MR, Dubois V, Claessens F, Verschueren SMP, Vanderschueren D, Gielen E, Jardí F. Muscle-bone interactions: From experimental models to the clinic? A critical update. Mol Cell Endocrinol 2016; 432:14-36. [PMID: 26506009 DOI: 10.1016/j.mce.2015.10.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
Bone is a biomechanical tissue shaped by forces from muscles and gravitation. Simultaneous bone and muscle decay and dysfunction (osteosarcopenia or sarco-osteoporosis) is seen in ageing, numerous clinical situations including after stroke or paralysis, in neuromuscular dystrophies, glucocorticoid excess, or in association with vitamin D, growth hormone/insulin like growth factor or sex steroid deficiency, as well as in spaceflight. Physical exercise may be beneficial in these situations, but further work is still needed to translate acceptable and effective biomechanical interventions like vibration therapy from animal models to humans. Novel antiresorptive and anabolic therapies are emerging for osteoporosis as well as drugs for sarcopenia, cancer cachexia or muscle wasting disorders, including antibodies against myostatin or activin receptor type IIA and IIB (e.g. bimagrumab). Ideally, increasing muscle mass would increase muscle strength and restore bone loss from disuse. However, the classical view that muscle is unidirectionally dominant over bone via mechanical loading is overly simplistic. Indeed, recent studies indicate a role for neuronal regulation of not only muscle but also bone metabolism, bone signaling pathways like receptor activator of nuclear factor kappa-B ligand (RANKL) implicated in muscle biology, myokines affecting bone and possible bone-to-muscle communication. Moreover, pharmacological strategies inducing isolated myocyte hypertrophy may not translate into increased muscle power because tendons, connective tissue, neurons and energy metabolism need to adapt as well. We aim here to critically review key musculoskeletal molecular pathways involved in mechanoregulation and their effect on the bone-muscle unit as a whole, as well as preclinical and emerging clinical evidence regarding the effects of sarcopenia therapies on osteoporosis and vice versa.
Collapse
Affiliation(s)
- Michaël R Laurent
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium.
| | - Vanessa Dubois
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Sabine M P Verschueren
- Research Group for Musculoskeletal Rehabilitation, Department of Rehabilitation Science, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Evelien Gielen
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
75
|
Seref-Ferlengez Z, Suadicani SO, Thi MM. A new perspective on mechanisms governing skeletal complications in type 1 diabetes. Ann N Y Acad Sci 2016; 1383:67-79. [PMID: 27571221 DOI: 10.1111/nyas.13202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/11/2016] [Accepted: 07/18/2016] [Indexed: 12/29/2022]
Abstract
This review focuses on bone mechanobiology in type 1 diabetes (T1D), an area of research on diabetes-associated skeletal complications that is still in its infancy. We first provide a brief overview of the deleterious effects of diabetes on the skeleton and of the knowledge gained from studies with rodent models of T1D. Second, we discuss two specific hallmarks of T1D, low insulin and high glucose, and address the extent to which they affect skeletal health. Third, we highlight the mechanosensitive nature of bone tissue and the importance of mechanical loading for bone health. We also summarize recent advances in bone mechanobiology that implicate osteocytes as the mechanosensors and major regulatory cells in the bone. Finally, we discuss recent evidence indicating that the diabetic bone is "deaf" to mechanical loading and that osteocytes are central players in mechanisms that lead to bone loss in T1D.
Collapse
Affiliation(s)
- Zeynep Seref-Ferlengez
- Department of Orthopaedic Surgery.,Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE)
| | - Sylvia O Suadicani
- Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE).,Department of Neuroscience.,Department of Urology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
| | - Mia M Thi
- Department of Orthopaedic Surgery.,Laboratories of Musculoskeletal Orthopedic Research at Einstein-Montefiore (MORE).,Department of Neuroscience
| |
Collapse
|
76
|
Liu C, Cui X, Ackermann TM, Flamini V, Chen W, Castillo AB. Osteoblast-derived paracrine factors regulate angiogenesis in response to mechanical stimulation. Integr Biol (Camb) 2016; 8:785-94. [PMID: 27332785 PMCID: PMC8274385 DOI: 10.1039/c6ib00070c] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Angiogenesis is a process by which new blood vessels emerge from existing vessels through endothelial cell sprouting, migration, proliferation, and tubule formation. Angiogenesis during skeletal growth, homeostasis and repair is a complex and incompletely understood process. As the skeleton adapts to mechanical loading, we hypothesized that mechanical stimulation regulates "osteo-angio" crosstalk in the context of angiogenesis. We showed that conditioned media (CM) from osteoblasts exposed to fluid shear stress enhanced endothelial cell proliferation and migration, but not tubule formation, relative to CM from static cultures. Endothelial cell sprouting was studied using a dual-channel collagen gel-based microfluidic device that mimics vessel geometry. Static CM enhanced endothelial cell sprouting frequency, whereas loaded CM significantly enhanced both frequency and length. Both sprouting frequency and length were significantly enhanced in response to factors released from osteoblasts exposed to fluid shear stress in an adjacent channel. Osteoblasts released angiogenic factors, of which osteopontin, PDGF-AA, IGBP-2, MCP-1, and Pentraxin-3 were upregulated in response to mechanical loading. These data suggest that in vivo mechanical forces regulate angiogenesis in bone by modulating "osteo-angio" crosstalk through release of paracrine factors, which we term "osteokines".
Collapse
Affiliation(s)
- Chao Liu
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA. and Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY 10003, USA
| | - Xin Cui
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA.
| | - Thomas M Ackermann
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA.
| | - Vittoria Flamini
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA.
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA.
| | - Alesha B Castillo
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA. and Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY 10003, USA
| |
Collapse
|
77
|
Pacheco-Costa R, Davis H, Atkinson E, Katchburian E, Plotkin L, Reginato R. Osteocytic connexin 43 is not required for the increase in bone mass induced by intermittent PTH administration in male mice. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2016; 16:45-57. [PMID: 26944823 PMCID: PMC5089455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate whether osteocytic connexin 43 (Cx43) is required for the bone response to intermittent PTH administration, and whether the connexin is involved in maintaining the bone matrix. METHODS Human PTH(1-34) was injected to adult male mice expressing (Cx43(fl/fl)) or not osteocytic Cx43 (Cx43(fl/fl);DMP1-8kb-Cre) daily (100 µg/kg/d) for 14 days. RESULTS Cx43(fl/fl);DMP1-8kb-Cre mice have no difference in body weight and BMD from 1 to 4 months of age. Intermittent PTH administration increased BMD and BV/TV and induced a similar increase in type I collagen, alkaline phosphatase, runx2, osteocalcin, and bone sialoprotein expression in mice from both genotypes. On the other hand, osteocytic deletion of Cx43 did not alter mRNA levels of glycosaminoglycans, proteoglycans, collagens and osteoblast-related genes. In addition, expression of collagens assessed by immunohistochemistry was not affected by deleting osteocytic Cx43. However, PTH administration increased type II collagen only in Cx43(fl/fl) control mice, whereas hormone increased type I collagen expression only in Cx43(fl/fl);DMP1-8kb-Cre mice. Furthermore, PTH increased maturity of collagen fibers in control, but not in Cx43-deficient mice. CONCLUSION Expression of Cx43 in osteocytes is dispensable for bone anabolism induced by intermittent PTH administration; but it can modulate, at least in part, the effect of PTH on the bone matrix environment.
Collapse
Affiliation(s)
- R. Pacheco-Costa
- Department of Morphology & Genetics, Federal University of São Paulo School of Medicine, São Paulo, SP 04023-900, Brazil,Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - H.M. Davis
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - E.G. Atkinson
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - E. Katchburian
- Department of Morphology & Genetics, Federal University of São Paulo School of Medicine, São Paulo, SP 04023-900, Brazil
| | - L.I. Plotkin
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - R.D. Reginato
- Department of Morphology & Genetics, Federal University of São Paulo School of Medicine, São Paulo, SP 04023-900, Brazil,Corresponding author: Rejane D. Reginato, Ph.D., Mineralized Tissue and Histology Research Laboratory, Department of Morphology and Genetics, Federal Universiy of São Paulo School of Medicine, Rua Botucatu, 740. Ed. Lemos Torres, São Paulo, SP 04023-900, Brazil E-mail:
| |
Collapse
|
78
|
Berke IM, Miola JP, David MA, Smith MK, Price C. Seeing through Musculoskeletal Tissues: Improving In Situ Imaging of Bone and the Lacunar Canalicular System through Optical Clearing. PLoS One 2016; 11:e0150268. [PMID: 26930293 PMCID: PMC4773178 DOI: 10.1371/journal.pone.0150268] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 02/11/2016] [Indexed: 12/17/2022] Open
Abstract
In situ, cells of the musculoskeletal system reside within complex and often interconnected 3-D environments. Key to better understanding how 3-D tissue and cellular environments regulate musculoskeletal physiology, homeostasis, and health is the use of robust methodologies for directly visualizing cell-cell and cell-matrix architecture in situ. However, the use of standard optical imaging techniques is often of limited utility in deep imaging of intact musculoskeletal tissues due to the highly scattering nature of biological tissues. Drawing inspiration from recent developments in the deep-tissue imaging field, we describe the application of immersion based optical clearing techniques, which utilize the principle of refractive index (RI) matching between the clearing/mounting media and tissue under observation, to improve the deep, in situ imaging of musculoskeletal tissues. To date, few optical clearing techniques have been applied specifically to musculoskeletal tissues, and a systematic comparison of the clearing ability of optical clearing agents in musculoskeletal tissues has yet to be fully demonstrated. In this study we tested the ability of eight different aqueous and non-aqueous clearing agents, with RIs ranging from 1.45 to 1.56, to optically clear murine knee joints and cortical bone. We demonstrated and quantified the ability of these optical clearing agents to clear musculoskeletal tissues and improve both macro- and micro-scale imaging of musculoskeletal tissue across several imaging modalities (stereomicroscopy, spectroscopy, and one-, and two-photon confocal microscopy) and investigational techniques (dynamic bone labeling and en bloc tissue staining). Based upon these findings we believe that optical clearing, in combination with advanced imaging techniques, has the potential to complement classical musculoskeletal analysis techniques; opening the door for improved in situ investigation and quantification of musculoskeletal tissues.
Collapse
Affiliation(s)
- Ian M. Berke
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, United States of America
| | - Joseph P. Miola
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, United States of America
| | - Michael A. David
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, United States of America
| | - Melanie K. Smith
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, United States of America
| | - Christopher Price
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, United States of America
- * E-mail:
| |
Collapse
|
79
|
Wijeratne SS, Martinez JR, Grindel BJ, Frey EW, Li J, Wang L, Farach-Carson MC, Kiang CH. Single molecule force measurements of perlecan/HSPG2: A key component of the osteocyte pericellular matrix. Matrix Biol 2015; 50:27-38. [PMID: 26546708 DOI: 10.1016/j.matbio.2015.11.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/01/2015] [Accepted: 11/02/2015] [Indexed: 12/30/2022]
Abstract
Perlecan/HSPG2, a large, monomeric heparan sulfate proteoglycan (HSPG), is a key component of the lacunar canalicular system (LCS) of cortical bone, where it is part of the mechanosensing pericellular matrix (PCM) surrounding the osteocytic processes and serves as a tethering element that connects the osteocyte cell body to the bone matrix. Within the pericellular space surrounding the osteocyte cell body, perlecan can experience physiological fluid flow drag force and in that capacity function as a sensor to relay external stimuli to the osteocyte cell membrane. We previously showed that a reduction in perlecan secretion alters the PCM fiber composition and interferes with bone's response to a mechanical loading in vivo. To test our hypothesis that perlecan core protein can sustain tensile forces without unfolding under physiological loading conditions, atomic force microscopy (AFM) was used to capture images of perlecan monomers at nanoscale resolution and to perform single molecule force measurement (SMFMs). We found that the core protein of purified full-length human perlecan is of suitable size to span the pericellular space of the LCS, with a measured end-to-end length of 170±20 nm and a diameter of 2-4 nm. Force pulling revealed a strong protein core that can withstand over 100 pN of tension well over the drag forces that are estimated to be exerted on the individual osteocyte tethers. Data fitting with an extensible worm-like chain model showed that the perlecan protein core has a mean elastic constant of 890 pN and a corresponding Young's modulus of 71 MPa. We conclude that perlecan has physical properties that would allow it to act as a strong but elastic tether in the LCS.
Collapse
Affiliation(s)
- Sithara S Wijeratne
- Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA
| | | | - Brian J Grindel
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Eric W Frey
- Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA
| | - Jingqiang Li
- Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Mary C Farach-Carson
- Department of BioSciences, Rice University, Houston, TX 77005, USA; Department of Bioengineering, Rice University, Houston, TX 77005, USA.
| | - Ching-Hwa Kiang
- Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA; Department of Bioengineering, Rice University, Houston, TX 77005, USA
| |
Collapse
|
80
|
Lai X, Price C, Modla S, Thompson WR, Caplan J, Kirn-Safran CB, Wang L. The dependences of osteocyte network on bone compartment, age, and disease. Bone Res 2015; 3. [PMID: 26213632 PMCID: PMC4511381 DOI: 10.1038/boneres.2015.9] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Osteocytes, the most abundant bone cells, form an interconnected network in the lacunar-canalicular pore system (LCS) buried within the mineralized matrix, which allows osteocytes to obtain nutrients from the blood supply, sense external mechanical signals, and communicate among themselves and with other cells on bone surfaces. In this study, we examined key features of the LCS network including the topological parameter and the detailed structure of individual connections and their variations in cortical and cancellous compartments, at different ages, and in two disease conditions with altered mechanosensing (perlecan deficiency and diabetes). LCS network showed both topological stability, in terms of conservation of connectivity among osteocyte lacunae (similar to the "nodes" in a computer network), and considerable variability the pericellular annular fluid gap surrounding lacunae and canaliculi (similar to the "bandwidth" of individual links in a computer network). Age, in the range of our study (15-32 weeks), affected only the pericellular fluid annulus in cortical bone but not in cancellous bone. Diabetes impacted the spacing of the lacunae, while the perlecan deficiency had a profound influence on the pericellular fluid annulus. The LCS network features play important roles in osteocyte signaling and regulation of bone growth and adaptation.
Collapse
Affiliation(s)
- Xiaohan Lai
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Christopher Price
- Biomedical Engineering Program, University of Delaware, Newark, DE, USA
| | - Shannon Modla
- DBI Bioimaging Center, University of Delaware, Newark, DE, USA
| | - William R Thompson
- Department of Physical Therapy, Indiana University, Indianapolis, IN, USA
| | - Jeffrey Caplan
- DBI Bioimaging Center, University of Delaware, Newark, DE, USA
| | | | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|
81
|
Aiello BR, Iriarte-Diaz J, Blob RW, Butcher MT, Carrano MT, Espinoza NR, Main RP, Ross CF. Bone strain magnitude is correlated with bone strain rate in tetrapods: implications for models of mechanotransduction. Proc Biol Sci 2015; 282:20150321. [PMID: 26063842 PMCID: PMC4590470 DOI: 10.1098/rspb.2015.0321] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/13/2015] [Indexed: 11/12/2022] Open
Abstract
Hypotheses suggest that structural integrity of vertebrate bones is maintained by controlling bone strain magnitude via adaptive modelling in response to mechanical stimuli. Increased tissue-level strain magnitude and rate have both been identified as potent stimuli leading to increased bone formation. Mechanotransduction models hypothesize that osteocytes sense bone deformation by detecting fluid flow-induced drag in the bone's lacunar-canalicular porosity. This model suggests that the osteocyte's intracellular response depends on fluid-flow rate, a product of bone strain rate and gradient, but does not provide a mechanism for detection of strain magnitude. Such a mechanism is necessary for bone modelling to adapt to loads, because strain magnitude is an important determinant of skeletal fracture. Using strain gauge data from the limb bones of amphibians, reptiles, birds and mammals, we identified strong correlations between strain rate and magnitude across clades employing diverse locomotor styles and degrees of rhythmicity. The breadth of our sample suggests that this pattern is likely to be a common feature of tetrapod bone loading. Moreover, finding that bone strain magnitude is encoded in strain rate at the tissue level is consistent with the hypothesis that it might be encoded in fluid-flow rate at the cellular level, facilitating bone adaptation via mechanotransduction.
Collapse
Affiliation(s)
- B R Aiello
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA
| | - J Iriarte-Diaz
- Department of Oral Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - R W Blob
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - M T Butcher
- Department of Biological Sciences, Youngstown State University, Youngstown, OH 44555, USA
| | - M T Carrano
- Department of Paleobiology, Smithsonian Institution, Washington, DC 20013, USA
| | - N R Espinoza
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - R P Main
- Department of Basic Medical Sciences, College of Veterinary Medicine and Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - C F Ross
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
82
|
Osteocyte specific responses to soluble and mechanical stimuli in a stem cell derived culture model. Sci Rep 2015; 5:11049. [PMID: 26056071 PMCID: PMC4460727 DOI: 10.1038/srep11049] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 05/07/2015] [Indexed: 11/08/2022] Open
Abstract
Studying osteocyte behavior in culture has proven difficult because these embedded cells require spatially coordinated interactions with the matrix and surrounding cells to achieve the osteocyte phenotype. Using an easily attainable source of bone marrow mesenchymal stem cells, we generated cells with the osteocyte phenotype within two weeks. These "stem cell derived-osteocytes" (SCD-O) displayed stellate morphology and lacunocanalicular ultrastructure. Osteocytic genes Sost, Dmp1, E11, and Fgf23 were maximally expressed at 15 days and responded to PTH and 1,25(OH)2D3. Production of sclerostin mRNA and protein, within 15 days of culture makes the SCD-O model ideal for elucidating regulatory mechanisms. We found sclerostin to be regulated by mechanical factors, where low intensity vibration significantly reduced Sost expression. Additionally, this model recapitulates sclerostin production in response to osteoactive hormones, as PTH or LIV repressed secretion of sclerostin, significantly impacting Wnt-mediated Axin2 expression, via β-catenin signaling. In summary, SCD-O cells produce abundant matrix, rapidly attain the osteocyte phenotype, and secrete functional factors including sclerostin under non-immortalized conditions. This culture model enables ex vivo observations of osteocyte behavior while preserving an organ-like environment. Furthermore, as marrow-derived mesenchymal stem cells can be obtained from transgenic animals; our model enables study of genetic control of osteocyte behaviors.
Collapse
|
83
|
Strain amplification analysis of an osteocyte under static and cyclic loading: a finite element study. BIOMED RESEARCH INTERNATIONAL 2015; 2015:376474. [PMID: 25664319 PMCID: PMC4312579 DOI: 10.1155/2015/376474] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/21/2014] [Accepted: 09/25/2014] [Indexed: 12/22/2022]
Abstract
Osteocytes, the major type of bone cells which reside in their lacunar and canalicular system within the bone matrix, function as biomechanosensors and biomechanotransducers of the bone. Although biomechanical behaviour of the osteocyte-lacunar-canalicular system has been investigated in previous studies mostly using computational 2-dimensional (2D) geometric models, only a few studies have used the 3-dimensional (3D) finite element (FE) model. In the current study, a 3D FE model was used to predict the responses of strain distributions of osteocyte-lacunar-canalicular system analyzed under static and cyclic loads. The strain amplification factor was calculated for all simulations. Effects on the strain of the osteocyte system were investigated under 500, 1500, 2000, and 3000 microstrain loading magnitudes and 1, 5, 10, 40, and 100 Hz loading frequencies. The maximum strain was found to change with loading magnitude and frequency. It was observed that maximum strain under 3000-microstrain loading was higher than those under 500, 1500, and 2000 microstrains. When the loading strain reached the maximum magnitude, the strain amplification factor of 100 Hz was higher than those of the other frequencies. Data from this 3D FE model study suggests that the strain amplification factor of the osteocyte-lacunar-canalicular system increases with loading frequency and loading strain increasing.
Collapse
|
84
|
Lowe DA, Lepori-Bui N, Fomin PV, Sloofman LG, Zhou X, Farach-Carson MC, Wang L, Kirn-Safran CB. Deficiency in perlecan/HSPG2 during bone development enhances osteogenesis and decreases quality of adult bone in mice. Calcif Tissue Int 2014; 95:29-38. [PMID: 24798737 PMCID: PMC4137566 DOI: 10.1007/s00223-014-9859-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 04/10/2014] [Indexed: 12/22/2022]
Abstract
Perlecan/HSPG2 (Pln) is a large heparan sulfate proteoglycan abundant in the extracellular matrix of cartilage and the lacunocanalicular space of adult bones. Although Pln function during cartilage development is critical, evidenced by deficiency disorders including Schwartz-Jampel Syndrome and dyssegmental dysplasia Silverman-Handmaker type, little is known about its function in development of bone shape and quality. The purpose of this study was to understand the contribution of Pln to bone geometric and mechanical properties. We used hypomorph mutant mice that secrete negligible amount of Pln into skeletal tissues and analyzed their adult bone properties using micro-computed tomography and three-point-bending tests. Bone shortening and widening in Pln mutants was observed and could be attributed to loss of growth plate organization and accelerated osteogenesis that was reflected by elevated cortical thickness at older ages. This effect was more pronounced in Pln mutant females, indicating a sex-specific effect of Pln deficiency on bone geometry. Additionally, mutant females, and to a lesser extent mutant males, increased their elastic modulus and bone mineral densities to counteract changes in bone shape, but at the expense of increased brittleness. In summary, Pln deficiency alters cartilage matrix patterning and, as we now show, coordinately influences bone formation and calcification.
Collapse
Affiliation(s)
- Dylan A. Lowe
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Nadia Lepori-Bui
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Peter V. Fomin
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Laura G. Sloofman
- University of Delaware, Department of Biological Sciences, Newark, DE
| | - Xiaozhou Zhou
- University of Delaware, Department of Mechanical Engineering, Newark, DE
| | - Mary C. Farach-Carson
- University of Delaware, Department of Biological Sciences, Newark, DE
- Rice University, Department of Biochemistry and Cell Biology, Houston, TX
| | - Liyun Wang
- University of Delaware, Department of Mechanical Engineering, Newark, DE
| | - Catherine B. Kirn-Safran
- University of Delaware, Department of Biological Sciences, Newark, DE
- author to whom correspondence should be addressed: Catherine Kirn-Safran, University of Delaware, Department of Biological Science, 310 Wolf Hall, Newark, DE 19716, Tel: (302) 831-3249, Fax: (302) 831-2281,
| |
Collapse
|