1
|
Chen N, Danalache M, Liang C, Alexander D, Umrath F. Mechanosignaling in Osteoporosis: When Cells Feel the Force. Int J Mol Sci 2025; 26:4007. [PMID: 40362247 PMCID: PMC12071322 DOI: 10.3390/ijms26094007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Bone is a highly mechanosensitive tissue, where mechanical signaling plays a central role in maintaining skeletal homeostasis. Mechanotransduction regulates the balance between bone formation and resorption through coordinated interactions among bone cells. Key mechanosensing structures-including the extracellular/pericellular matrix (ECM/PCM), integrins, ion channels, connexins, and primary cilia, translate mechanical cues into biochemical signals that drive bone adaptation. Disruptions in mechanotransduction are increasingly recognized as an important factor in osteoporosis. Under pathological conditions, impaired mechanical signaling reduces bone formation and accelerates bone resorption, leading to skeletal fragility. Defects in mechanotransduction disrupt key pathways involved in bone metabolism, further exacerbating bone loss. Therefore, targeting mechanotransduction presents a promising pharmacological strategy for osteoporosis treatment. Recent advances have focused on developing drugs that enhance bone mechanosensitivity by modulating key mechanotransduction pathways, including integrins, ion channels, connexins, and Wnt signaling. A deeper understanding of mechanosignaling mechanisms may pave the way for novel therapeutic approaches aimed at restoring bone mass, mechanical integrity, and mechanosensitive bone adaptation.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
| | - Marina Danalache
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
| | - Chen Liang
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
| | - Dorothea Alexander
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Felix Umrath
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
2
|
Melrose J. Glycosaminoglycans, Instructive Biomolecules That Regulate Cellular Activity and Synaptic Neuronal Control of Specific Tissue Functional Properties. Int J Mol Sci 2025; 26:2554. [PMID: 40141196 PMCID: PMC11942259 DOI: 10.3390/ijms26062554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Glycosaminoglycans (GAGs) are a diverse family of ancient biomolecules that evolved over millennia as key components in the glycocalyx that surrounds all cells. GAGs have molecular recognition and cell instructive properties when attached to cell surface and extracellular matrix (ECM) proteoglycans (PGs), which act as effector molecules that regulate cellular behavior. The perception of mechanical cues which arise from perturbations in the ECM microenvironment allow the cell to undertake appropriate biosynthetic responses to maintain ECM composition and tissue function. ECM PGs substituted with GAGs provide structural support to weight-bearing tissues and an ability to withstand shear forces in some tissue contexts. This review outlines the structural complexity of GAGs and the diverse functional properties they convey to cellular and ECM PGs. PGs have important roles in cartilaginous weight-bearing tissues and fibrocartilages subject to tension and high shear forces and also have important roles in vascular and neural tissues. Specific PGs have roles in synaptic stabilization and convey specificity and plasticity in the regulation of neurophysiological responses in the CNS/PNS that control tissue function. A better understanding of GAG instructional roles over cellular behavior may be insightful for the development of GAG-based biotherapeutics designed to treat tissue dysfunction in disease processes and in novel tissue repair strategies following trauma. GAGs have a significant level of sophistication over the control of cellular behavior in many tissue contexts, which needs to be fully deciphered in order to achieve a useful therapeutic product. GAG biotherapeutics offers exciting opportunities in the modern glycomics arena.
Collapse
Affiliation(s)
- James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia;
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School, Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
3
|
Hopkinson M, Pitsillides AA. Extracellular matrix: Dystroglycan interactions-Roles for the dystrophin-associated glycoprotein complex in skeletal tissue dynamics. Int J Exp Pathol 2025; 106:e12525. [PMID: 39923120 PMCID: PMC11807010 DOI: 10.1111/iep.12525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/23/2024] [Accepted: 12/29/2024] [Indexed: 02/10/2025] Open
Abstract
Contributions made by the dystrophin-associated glycoprotein complex (DGC) to cell-cell and cell-extracellular matrix (ECM) interactions are vital in development, homeostasis and pathobiology. This review explores how DGC functions may extend to skeletal pathophysiology by appraising the known roles of its major ECM ligands, and likely associated DGC signalling pathways, in regulating cartilage and bone cell behaviour and emergent skeletal phenotypes. These considerations will be contextualised by highlighting the potential of studies into the role of the DGC in isolated chondrocytes, osteoblasts and osteoclasts, and by fuller deliberation of skeletal phenotypes that may emerge in very young mice lacking vital, yet diverse core elements of the DGC. Our review points to roles for individual DGC components-including the glycosylation of dystroglycan itself-beyond the establishment of membrane stability which clearly accounts for severe muscle phenotypes in muscular dystrophy. It implies that the short stature, low bone mineral density, poor bone health and greater fracture risk in these patients, which has been attributed due to primary deficiencies in muscle-evoked skeletal loading, may instead arise due to primary roles for the DGC in controlling skeletal tissue (re)modelling.
Collapse
Affiliation(s)
- Mark Hopkinson
- Skeletal Biology Group, Comparative Biomedical SciencesRoyal Veterinary CollegeLondonUK
| | - Andrew A. Pitsillides
- Skeletal Biology Group, Comparative Biomedical SciencesRoyal Veterinary CollegeLondonUK
| |
Collapse
|
4
|
Baumann BT, Nieuwstraten J, Konrads C, Guilak F, Danalache M. Cracking the Pericellular Matrix Code: Exploring how MMP-2, -3, and -7 influence matrix breakdown and biomechanical properties. Osteoarthritis Cartilage 2025; 33:241-246. [PMID: 39322008 DOI: 10.1016/j.joca.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The intricate process of articular cartilage remodeling, pivotal for both physiological functions and osteoarthritis (OA) progression, is orchestrated through a balance of matrix synthesis and breakdown, which is mediated by matrix metalloproteinase enzymes (MMPs). At the heart of this remodeling lies the pericellular matrix (PCM), a specialized microenvironment encapsulating each chondrocyte and composed mainly of collagen type VI and perlecan. The aim of this study was to assess the impact of MMP-2, -3, and -7 on the structural integrity and biomechanical attributes of the PCM. METHODS Human articular cartilage explants (N = 10 patients) were incubated with activated MMP-2, -3, or -7, individually or in combination. Structural alterations in the PCM were evaluated by immunolabeling. The biomechanical properties of the PCM were measured using atomic force microscopy (AFM). RESULTS Collagen type VI structural integrity and fluorescence intensity uniformly decreased across all enzyme groups, while perlecan was selectively affected by MMP-3 and -7. AFM measurements demonstrated decreased PCM stiffness after incubation with individual MMPs, leading to an overall ∼31% reduction in elastic modulus for each enzyme. Combinations of enzymes induced comparable significant biomechanical alterations (∼35%), except for MMP-2+MMP-7. DISCUSSION This study highlights the significant influence of MMP-induced alterations in PCM composition on biomechanical properties, mirroring characteristics observed in early OA. Each MMP showed specificity in breaking down PCM, and an intriguing interplay, especially between MMP-2 and -7, indicated reduced efficacy in lowering PCM stiffness. Overall, MMP-2, -3, and -7 directly induce functional and structural PCM modifications.
Collapse
Affiliation(s)
- Benjamin Tizian Baumann
- Department of Orthopedic Surgery, University of Tübingen, Tübingen, Germany; Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Jule Nieuwstraten
- Department of Orthopedic Surgery, University of Tübingen, Tübingen, Germany; Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Christian Konrads
- Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany; Department of Orthopaedics and Traumatology, Hanseatic Hospital Stralsund, Stralsund, Germany.
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA.
| | - Marina Danalache
- Department of Orthopedic Surgery, University of Tübingen, Tübingen, Germany; Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Guerra RM, Fowler VM, Wang L. Osteocyte Dendrites: How Do They Grow, Mature, and Degenerate in Mineralized Bone? Cytoskeleton (Hoboken) 2024. [PMID: 39651620 DOI: 10.1002/cm.21964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024]
Abstract
Osteocytes, the most abundant bone cells, form an extensive cellular network via interconnecting dendrites. Like neurons in the brain, the long-lived osteocytes perceive mechanical and biological inputs and signal to other effector cells, leading to the homeostasis and turnover of bone tissues. Despite the appreciation of osteocytes' vital roles in bone biology, the initiation, growth, maintenance, and eventual degradation of osteocyte dendrites are poorly understood due to their full encasement by mineralized matrix. With the advancement of imaging modalities and genetic models, the architectural organization and molecular composition of the osteocyte dendrites, as well as their morphological changes with aging and diseases, have begun to be revealed. However, several long-standing mysteries remain unsolved, including (1) how the dendrites are initiated and elongated when a surface osteoblast becomes embedded as an osteocyte; (2) how the dendrites maintain a relatively stable morphology during their decades-long life span; (3) what biological processes control the dendrite morphology, connectivity, and stability; and (4) if these processes are influenced by age, sex, hormones, and mechanical loading. Our review of long, thin actin filament (F-actin)-containing processes extending from other cells leads to a working model that serves as a starting point to investigate the formation and maintenance of osteocyte dendrites and their degradation with aging and diseases.
Collapse
Affiliation(s)
- Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Velia M Fowler
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Liyun Wang
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
6
|
Melrose J, Guilak F. Diverse and multifunctional roles for perlecan ( HSPG2) in repair of the intervertebral disc. JOR Spine 2024; 7:e1362. [PMID: 39081381 PMCID: PMC11286675 DOI: 10.1002/jsp2.1362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Perlecan is a widely distributed, modular, and multifunctional heparan sulfate proteoglycan, which facilitates cellular communication with the extracellular environment to promote tissue development, tissue homeostasis, and optimization of biomechanical tissue functions. Perlecan-mediated osmotic mechanotransduction serves to regulate the metabolic activity of cells in tissues subjected to tension, compression, or shear. Perlecan interacts with a vast array of extracellular matrix (ECM) proteins through which it stabilizes tissues and regulates the proliferation or differentiation of resident cell populations. Here we examine the roles of the HS-proteoglycan perlecan in the normal and destabilized intervertebral disc. The intervertebral disc cell has evolved to survive in a hostile weight bearing, acidic, low oxygen tension, and low nutrition environment, and perlecan provides cytoprotection, shields disc cells from excessive compressive forces, and sequesters a range of growth factors in the disc cell environment where they aid in cellular survival, proliferation, and differentiation. The cells in mechanically destabilized connective tissues attempt to re-establish optimal tissue composition and tissue functional properties by changing the properties of their ECM, in the process of chondroid metaplasia. We explore the possibility that perlecan assists in these cell-mediated tissue remodeling responses by regulating disc cell anabolism. Perlecan's mechano-osmotic transductive property may be of potential therapeutic application.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling InstituteNorthern Sydney Local Health DistrictSt. LeonardsNew South WalesAustralia
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNew South WalesAustralia
- Sydney Medical School, NorthernThe University of SydneySt. LeonardsNew South WalesAustralia
- Faculty of Medicine and HealthThe University of Sydney, Royal North Shore HospitalSt. LeonardsNew South WalesAustralia
| | - Farshid Guilak
- Department of Orthopaedic SurgeryWashington UniversitySt. LouisMissouriUSA
- Department of OrthopaedicsShriners Hospitals for ChildrenSt. LouisMissouriUSA
| |
Collapse
|
7
|
Melrose J. Dystroglycan-HSPG interactions provide synaptic plasticity and specificity. Glycobiology 2024; 34:cwae051. [PMID: 39223703 PMCID: PMC11368572 DOI: 10.1093/glycob/cwae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
AIM This study examined the roles of the laminin and proteoglycan receptor dystroglycan (DG) in extracellular matrix stabilization and cellular mechanosensory processes conveyed through communication between the extracellular matrix (ECM) and cytoskeleton facilitated by DG. Specific functional attributes of HS-proteoglycans (HSPGs) are conveyed through interactions with DG and provide synaptic specificity through diverse interactions with an extensive range of cell attachment and adaptor proteins which convey synaptic plasticity. HSPG-DG interactions are important in phototransduction and neurotransduction and facilitate retinal bipolar-photoreceptor neuronal signaling in vision. Besides synaptic stabilization, HSPG-DG interactions also stabilize basement membranes and the ECM and have specific roles in the assembly and function of the neuromuscular junction. This provides neuromuscular control of muscle systems that control conscious body movement as well as essential autonomic control of diaphragm, intercostal and abdominal muscles and muscle systems in the face, mouth and pharynx which assist in breathing processes. DG is thus a multifunctional cell regulatory glycoprotein receptor and regulates a diverse range of biological and physiological processes throughout the human body. The unique glycosylation of the αDG domain is responsible for its diverse interactions with ECM components in cell-ECM signaling. Cytoskeletal cell regulatory switches assembled by the βDG domain in its role as a nuclear scaffolding protein respond to such ECM cues to regulate cellular behavior and tissue homeostasis thus DG has fascinating and diverse roles in health and disease.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, St. Leonards, NSW 2065, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
8
|
Mongiat M, Pascal G, Poletto E, Williams DM, Iozzo RV. Proteoglycans of basement membranes: Crucial controllers of angiogenesis, neurogenesis, and autophagy. PROTEOGLYCAN RESEARCH 2024; 2:e22. [PMID: 39184370 PMCID: PMC11340296 DOI: 10.1002/pgr2.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/02/2024] [Indexed: 08/27/2024]
Abstract
Anti-angiogenic therapy is an established method for the treatment of several cancers and vascular-related diseases. Most of the agents employed target the vascular endothelial growth factor A, the major cytokine stimulating angiogenesis. However, the efficacy of these treatments is limited by the onset of drug resistance. Therefore, it is of fundamental importance to better understand the mechanisms that regulate angiogenesis and the microenvironmental cues that play significant role and influence patient treatment and outcome. In this context, here we review the importance of the three basement membrane heparan sulfate proteoglycans (HSPGs), namely perlecan, agrin and collagen XVIII. These HSPGs are abundantly expressed in the vasculature and, due to their complex molecular architecture, they interact with multiple endothelial cell receptors, deeply affecting their function. Under normal conditions, these proteoglycans exert pro-angiogenic functions. However, in pathological conditions such as cancer and inflammation, extracellular matrix remodeling leads to the degradation of these large precursor molecules and the liberation of bioactive processed fragments displaying potent angiostatic activity. These unexpected functions have been demonstrated for the C-terminal fragments of perlecan and collagen XVIII, endorepellin and endostatin. These bioactive fragments can also induce autophagy in vascular endothelial cells which contributes to angiostasis. Overall, basement membrane proteoglycans deeply affect angiogenesis counterbalancing pro-angiogenic signals during tumor progression, and represent possible means to develop new prognostic biomarkers and novel therapeutic approaches for the treatment of solid tumors.
Collapse
Affiliation(s)
- Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Gabriel Pascal
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Davion M. Williams
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
9
|
Wang C, Fan M, Heo SJ, Adams SM, Li T, Liu Y, Li Q, Loebel C, Alisafaei F, Burdick JA, Lu XL, Birk DE, Mauck RL, Han L. Structure-Mechanics Principles and Mechanobiology of Fibrocartilage Pericellular Matrix: A Pivotal Role of Type V Collagen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600498. [PMID: 38979323 PMCID: PMC11230444 DOI: 10.1101/2024.06.26.600498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding resident cells in various tissue types, regulating matrix turnover, cell-matrix cross-talk and disease initiation. This study elucidated the structure-mechanical properties and mechanobiological functions of the PCM in fibrocartilage, a family of connective tissues that sustain complex tensile and compressive loads in vivo. Studying the murine meniscus as the model tissue, we showed that fibrocartilage PCM contains thinner, random collagen fibrillar networks that entrap proteoglycans, a structure distinct from the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). In comparison to the ECM, the PCM has a lower modulus and greater isotropy, but similar relative viscoelastic properties. In Col5a1 +/- menisci, the reduction of collagen V, a minor collagen localized in the PCM, resulted in aberrant fibril thickening with increased heterogeneity. Consequently, the PCM exhibited a reduced modulus, loss of isotropy and faster viscoelastic relaxation. This disrupted PCM contributes to perturbed mechanotransduction of resident meniscal cells, as illustrated by reduced intracellular calcium signaling, as well as upregulated biosynthesis of lysyl oxidase and tenascin C. When cultured in vitro, Col5a1 +/- meniscal cells synthesized a weakened nascent PCM, which had inferior properties towards protecting resident cells against applied tensile stretch. These findings underscore the PCM as a distinctive microstructure that governs fibrocartilage mechanobiology, and highlight the pivotal role of collagen V in PCM function. Targeting the PCM or its molecular constituents holds promise for enhancing not only meniscus regeneration and osteoarthritis intervention, but also addressing diseases across various fibrocartilaginous tissues.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Claudia Loebel
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, United States
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
10
|
Wright CS, Lewis KJ, Semon K, Yi X, Reyes Fernandez PC, Rust K, Prideaux M, Schneider A, Pederson M, Deosthale P, Plotkin LI, Hum JM, Sankar U, Farach-Carson MC, Robling AG, Thompson WR. Deletion of the auxiliary α2δ1 voltage sensitive calcium channel subunit in osteocytes and late-stage osteoblasts impairs femur strength and load-induced bone formation in male mice. J Bone Miner Res 2024; 39:298-314. [PMID: 38477790 DOI: 10.1093/jbmr/zjae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 12/03/2023] [Accepted: 12/27/2023] [Indexed: 03/14/2024]
Abstract
Osteocytes sense and respond to mechanical force by controlling the activity of other bone cells. However, the mechanisms by which osteocytes sense mechanical input and transmit biological signals remain unclear. Voltage-sensitive calcium channels (VSCCs) regulate calcium (Ca2+) influx in response to external stimuli. Inhibition or deletion of VSCCs impairs osteogenesis and skeletal responses to mechanical loading. VSCC activity is influenced by its auxiliary subunits, which bind the channel's α1 pore-forming subunit to alter intracellular Ca2+ concentrations. The α2δ1 auxiliary subunit associates with the pore-forming subunit via a glycosylphosphatidylinositol anchor and regulates the channel's calcium-gating kinetics. Knockdown of α2δ1 in osteocytes impairs responses to membrane stretch, and global deletion of α2δ1 in mice results in osteopenia and impaired skeletal responses to loading in vivo. Therefore, we hypothesized that the α2δ1 subunit functions as a mechanotransducer, and its deletion in osteocytes would impair skeletal development and load-induced bone formation. Mice (C57BL/6) with LoxP sequences flanking Cacna2d1, the gene encoding α2δ1, were crossed with mice expressing Cre under the control of the Dmp1 promoter (10 kb). Deletion of α2δ1 in osteocytes and late-stage osteoblasts decreased femoral bone quantity (P < .05) by DXA, reduced relative osteoid surface (P < .05), and altered osteoblast and osteocyte regulatory gene expression (P < .01). Cacna2d1f/f, Cre + male mice displayed decreased femoral strength and lower 10-wk cancellous bone in vivo micro-computed tomography measurements at the proximal tibia (P < .01) compared to controls, whereas Cacna2d1f/f, Cre + female mice showed impaired 20-wk cancellous and cortical bone ex vivo micro-computed tomography measurements (P < .05) vs controls. Deletion of α2δ1 in osteocytes and late-stage osteoblasts suppressed load-induced calcium signaling in vivo and decreased anabolic responses to mechanical loading in male mice, demonstrating decreased mechanosensitivity. Collectively, the α2δ1 auxiliary subunit is essential for the regulation of osteoid-formation, femur strength, and load-induced bone formation in male mice.
Collapse
Affiliation(s)
- Christian S Wright
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
| | - Karl J Lewis
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14850, United States
| | - Katelyn Semon
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN 46202, United States
- Department of Anatomy & Cell Biology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States
| | - Xin Yi
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
| | - Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
| | - Katie Rust
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN 46202, United States
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
| | - Artur Schneider
- Department of Physiology, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46202, United States
| | - Molly Pederson
- School of Science, Indiana University-Purdue University, Indianapolis, IN 46202, United States
| | - Padmini Deosthale
- Department of Anatomy & Cell Biology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States
| | - Lilian I Plotkin
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
- Department of Anatomy & Cell Biology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States
| | - Julia M Hum
- Department of Physiology, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46202, United States
| | - Uma Sankar
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
- Department of Anatomy & Cell Biology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas, Health Science Center, Houston, TX 78712, United States
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
- Department of Anatomy & Cell Biology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States
| | - William R Thompson
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, United States
- Department of Anatomy & Cell Biology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States
| |
Collapse
|
11
|
Farach-Carson MC, Wu D, França CM. Proteoglycans in Mechanobiology of Tissues and Organs: Normal Functions and Mechanopathology. PROTEOGLYCAN RESEARCH 2024; 2:e21. [PMID: 39584146 PMCID: PMC11584024 DOI: 10.1002/pgr2.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/03/2024] [Indexed: 11/26/2024]
Abstract
Proteoglycans (PGs) are a diverse class of glycoconjugates that serve critical functions in normal mechanobiology and mechanopathology. Both the protein cores and attached glycosaminoglycan (GAG) chains function in mechanically-sensitive processes, and loss of either can contribute to development of pathological conditions. PGs function as key components of the extracellular matrix (ECM) where they can serve as mechanosensors in mechanosensitive tissues including bone, cartilage, tendon, blood vessels and soft organs. The mechanical properties of these tissues depend on the presence and function of PGs, which play important roles in tissue elasticity, osmolarity and pressure sensing, and response to physical activity. Tissue responses depend on cell surface mechanoreceptors that include integrins, CD44, voltage sensitive ion channels, transient receptor potential (TRP) and piezo channels. PGs contribute to cell and molecular interplay in wound healing, fibrosis, and cancer, where they transduce the mechanical properties of the ECM and influence the progression of various context-specific conditions and diseases. The PGs that are most important in mechanobiology vary depending on the tissue and its functions and functional needs. Perlecan, for example, is important in the mechanobiology of basement membranes, cardiac and skeletal muscle, while aggrecan plays a primary role in the mechanical properties of cartilage and joints. A variety of techniques have been used to study the mechanobiology of PGs, including atomic force microscopy, mouse knockout models, and in vitro cell culture experiments with 3D organoid models. These studies have helped to elucidate the tissue-specific roles that PGs play in cell-level mechanosensing and tissue mechanics. Overall, the study of PGs in mechanobiology is yielding fundamental new concepts in the molecular basis of mechanosensing that can open the door to the development of new treatments for a host of conditions related to mechanopathology.
Collapse
Affiliation(s)
- Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX 77005
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX 77005
| | - Cristiane Miranda França
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, Portland, OR, 97201
- Knight Cancer Precision Biofabrication Hub, Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97201
| |
Collapse
|
12
|
Buck HV, Stains JP. Osteocyte-mediated mechanical response controls osteoblast differentiation and function. Front Physiol 2024; 15:1364694. [PMID: 38529481 PMCID: PMC10961341 DOI: 10.3389/fphys.2024.1364694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024] Open
Abstract
Low bone mass is a pervasive global health concern, with implications for osteoporosis, frailty, disability, and mortality. Lifestyle factors, including sedentary habits, metabolic dysfunction, and an aging population, contribute to the escalating prevalence of osteopenia and osteoporosis. The application of mechanical load to bone through physical activity and exercise prevents bone loss, while sufficient mechanical load stimulates new bone mass acquisition. Osteocytes, cells embedded within the bone, receive mechanical signals and translate these mechanical cues into biological signals, termed mechano-transduction. Mechano-transduction signals regulate other bone resident cells, such as osteoblasts and osteoclasts, to orchestrate changes in bone mass. This review explores the mechanisms through which osteocyte-mediated response to mechanical loading regulates osteoblast differentiation and bone formation. An overview of bone cell biology and the impact of mechanical load will be provided, with emphasis on the mechanical cues, mechano-transduction pathways, and factors that direct progenitor cells toward the osteoblast lineage. While there are a wide range of clinically available treatments for osteoporosis, the majority act through manipulation of the osteoclast and may have significant disadvantages. Despite the central role of osteoblasts to the deposition of new bone, few therapies directly target osteoblasts for the preservation of bone mass. Improved understanding of the mechanisms leading to osteoblastogenesis may reveal novel targets for translational investigation.
Collapse
Affiliation(s)
| | - Joseph Paul Stains
- School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
13
|
Melrose J. Hippo cell signaling and HS-proteoglycans regulate tissue form and function, age-dependent maturation, extracellular matrix remodeling, and repair. Am J Physiol Cell Physiol 2024; 326:C810-C828. [PMID: 38223931 DOI: 10.1152/ajpcell.00683.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
This review examined how Hippo cell signaling and heparan sulfate (HS)-proteoglycans (HSPGs) regulate tissue form and function. Despite being a nonweight-bearing tissue, the brain is regulated by Hippo mechanoresponsive cell signaling pathways during embryonic development. HS-proteoglycans interact with growth factors, morphogens, and extracellular matrix components to regulate development and pathology. Pikachurin and Eyes shut (Eys) interact with dystroglycan to stabilize the photoreceptor axoneme primary cilium and ribbon synapse facilitating phototransduction and neurotransduction with bipolar retinal neuronal networks in ocular vision, the primary human sense. Another HSPG, Neurexin interacts with structural and adaptor proteins to stabilize synapses and ensure specificity of neural interactions, and aids in synaptic potentiation and plasticity in neurotransduction. HSPGs also stabilize the blood-brain barrier and motor neuron basal structures in the neuromuscular junction. Agrin and perlecan localize acetylcholinesterase and its receptors in the neuromuscular junction essential for neuromuscular control. The primary cilium is a mechanosensory hub on neurons, utilized by YES associated protein (YAP)-transcriptional coactivator with PDZ-binding motif (TAZ) Hippo, Hh, Wnt, transforming growth factor (TGF)-β/bone matrix protein (BMP) receptor tyrosine kinase cell signaling. Members of the glypican HSPG proteoglycan family interact with Smoothened and Patched G-protein coupled receptors on the cilium to regulate Hh and Wnt signaling during neuronal development. Control of glycosyl sulfotransferases and endogenous protease expression by Hippo TAZ YAP represents a mechanism whereby the fine structure of HS-proteoglycans can be potentially modulated spatiotemporally to regulate tissue morphogenesis in a similar manner to how Hippo signaling controls sialyltransferase expression and mediation of cell-cell recognition, dysfunctional sialic acid expression is a feature of many tumors.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Sydney Medical School-Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Kelly MM, Sharma K, Wright CS, Yi X, Reyes Fernandez PC, Gegg AT, Gorrell TA, Noonan ML, Baghdady A, Sieger JA, Dolphin AC, Warden SJ, Deosthale P, Plotkin LI, Sankar U, Hum JM, Robling AG, Farach-Carson MC, Thompson WR. Loss of the auxiliary α 2δ 1 voltage-sensitive calcium channel subunit impairs bone formation and anabolic responses to mechanical loading. JBMR Plus 2024; 8:ziad008. [PMID: 38505532 PMCID: PMC10945727 DOI: 10.1093/jbmrpl/ziad008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/31/2023] [Accepted: 11/27/2023] [Indexed: 03/21/2024] Open
Abstract
Voltage-sensitive calcium channels (VSCCs) influence bone structure and function, including anabolic responses to mechanical loading. While the pore-forming (α1) subunit of VSCCs allows Ca2+ influx, auxiliary subunits regulate the biophysical properties of the pore. The α2δ1 subunit influences gating kinetics of the α1 pore and enables mechanically induced signaling in osteocytes; however, the skeletal function of α2δ1 in vivo remains unknown. In this work, we examined the skeletal consequences of deleting Cacna2d1, the gene encoding α2δ1. Dual-energy X-ray absorptiometry and microcomputed tomography imaging demonstrated that deletion of α2δ1 diminished bone mineral content and density in both male and female C57BL/6 mice. Structural differences manifested in both trabecular and cortical bone for males, while the absence of α2δ1 affected only cortical bone in female mice. Deletion of α2δ1 impaired skeletal mechanical properties in both sexes, as measured by three-point bending to failure. While no changes in osteoblast number or activity were found for either sex, male mice displayed a significant increase in osteoclast number, accompanied by increased eroded bone surface and upregulation of genes that regulate osteoclast differentiation. Deletion of α2δ1 also rendered the skeleton insensitive to exogenous mechanical loading in males. While previous work demonstrates that VSCCs are essential for anabolic responses to mechanical loading, the mechanism by which these channels sense and respond to force remained unclear. Our data demonstrate that the α2δ1 auxiliary VSCC subunit functions to maintain baseline bone mass and strength through regulation of osteoclast activity and also provides skeletal mechanotransduction in male mice. These data reveal a molecular player in our understanding of the mechanisms by which VSCCs influence skeletal adaptation.
Collapse
Affiliation(s)
- Madison M Kelly
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
- College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, United States
| | - Karan Sharma
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
- College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, United States
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
| | - Xin Yi
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
| | - Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
| | - Aaron T Gegg
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
| | - Taylor A Gorrell
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
| | - Megan L Noonan
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, United States
| | - Ahmed Baghdady
- College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, United States
| | - Jacob A Sieger
- College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, United States
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College of London, Gower Street, London WC1E 6BT, United Kingdom
| | - Stuart J Warden
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
- La Trobe Sport and Exercise Medicine Research Centre, La Trobe University, Melbourne Victoria 3086, DX 211319, Australia
| | - Padmini Deosthale
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
- Department of Anatomy, Cell Biology, & Physiology, Indiana University, Indianapolis, IN 46202, United States
| | - Lilian I Plotkin
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
- Department of Anatomy, Cell Biology, & Physiology, Indiana University, Indianapolis, IN 46202, United States
| | - Uma Sankar
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
- Department of Anatomy, Cell Biology, & Physiology, Indiana University, Indianapolis, IN 46202, United States
| | - Julia M Hum
- College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, United States
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
- Department of Anatomy, Cell Biology, & Physiology, Indiana University, Indianapolis, IN 46202, United States
| | - Mary C Farach-Carson
- Department of Diagnostic & Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77054, United States
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, United States
- College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, United States
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN 46202, United States
- Department of Anatomy, Cell Biology, & Physiology, Indiana University, Indianapolis, IN 46202, United States
| |
Collapse
|
15
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
16
|
Gao L, Liu G, Wu X, Liu C, Wang Y, Ma M, Ma Y, Hao Z. Osteocytes autophagy mediated by mTORC2 activation controls osteoblasts differentiation and osteoclasts activities under mechanical loading. Arch Biochem Biophys 2023; 742:109634. [PMID: 37164247 DOI: 10.1016/j.abb.2023.109634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/01/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Autophagy is an important mechanosensitive response for cellular homeostasis and survival in osteocytes. However, the mechanism and its effect on bone metabolism have not yet clarified. The objective of this study was to evaluate how compressive cyclic force (CCF) induced autophagic response in osteocytes and to determine the effect of mechanically induced-autophagy on bone cells including osteocytes, osteoblasts, and osteoclasts. Autophagic puncta observed in MLO-Y4 cells increased after exposure to CCF. The upregulated levels of the LC3-II isoform and the degradation of p62 further confirmed the increased autophagic flux. Additionally, ATP synthesis and release, osteocalcin (OCN) expression, and cell survival increased in osteocytes as well. The Murine osteoblasts MC3T3-E1 cells and RAW 264.7 macrophage cells were cultured in conditioned medium collected from MLO-Y4 cells subjected to CCF. The concentration of FGF23 increased and the concentrations of SOST and M-CSF and RANKL/OPG ratio decreased significantly in the conditioned medium. Moreover, the promotion of osteogenic differentiation in MC3T3-E1 cells and inhibition of osteoclastogenesis and function in RAW 264.7 cells were significantly attenuated when osteocytes autophagy was inhibited by siAtg7. Our findings suggested that CCF induced protective autophagy in osteocytes and subsequently enhanced osteocytes survival and osteoblasts differentiation and downregulated osteoclasts activities. Further study revealed that CCF induced autophagic response in osteocytes through mechanistic target of rapamycin complex 2 (mTORC2) activation. In conclusion, CCF-induced osteocytes autophagy upon mTORC2 activation promoted osteocytes survival and osteogenic response and decreased osteoclastic function. Thus, osteocytes autophagy will provide a promising target for better understanding of bone physiology and treatment of bone diseases.
Collapse
Affiliation(s)
- Li Gao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Gen Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Xiangnan Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Chuanzi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Yiqiao Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Meirui Ma
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Yuanyuan Ma
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China.
| | - Zhichao Hao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China.
| |
Collapse
|
17
|
Reyes Fernandez PC, Wright CS, Masterson AN, Yi X, Tellman TV, Bonteanu A, Rust K, Noonan ML, White KE, Lewis KJ, Sankar U, Hum JM, Bix G, Wu D, Robling AG, Sardar R, Farach-Carson MC, Thompson WR. Gabapentin Disrupts Binding of Perlecan to the α 2δ 1 Voltage Sensitive Calcium Channel Subunit and Impairs Skeletal Mechanosensation. Biomolecules 2022; 12:biom12121857. [PMID: 36551284 PMCID: PMC9776037 DOI: 10.3390/biom12121857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Our understanding of how osteocytes, the principal mechanosensors within bone, sense and perceive force remains unclear. Previous work identified "tethering elements" (TEs) spanning the pericellular space of osteocytes and transmitting mechanical information into biochemical signals. While we identified the heparan sulfate proteoglycan perlecan (PLN) as a component of these TEs, PLN must attach to the cell surface to induce biochemical responses. As voltage-sensitive calcium channels (VSCCs) are critical for bone mechanotransduction, we hypothesized that PLN binds the extracellular α2δ1 subunit of VSCCs to couple the bone matrix to the osteocyte membrane. Here, we showed co-localization of PLN and α2δ1 along osteocyte dendritic processes. Additionally, we quantified the molecular interactions between α2δ1 and PLN domains and demonstrated for the first time that α2δ1 strongly associates with PLN via its domain III. Furthermore, α2δ1 is the binding site for the commonly used pain drug, gabapentin (GBP), which is associated with adverse skeletal effects when used chronically. We found that GBP disrupts PLN::α2δ1 binding in vitro, and GBP treatment in vivo results in impaired bone mechanosensation. Our work identified a novel mechanosensory complex within osteocytes composed of PLN and α2δ1, necessary for bone force transmission and sensitive to the drug GBP.
Collapse
Affiliation(s)
- Perla C. Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Christian S. Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Adrianna N. Masterson
- Department of Chemistry and Chemical Biology, School of Science, Indiana University, Indianapolis, IN 46202, USA
| | - Xin Yi
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Tristen V. Tellman
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Andrei Bonteanu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - Katie Rust
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Megan L. Noonan
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Kenneth E. White
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Karl J. Lewis
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Uma Sankar
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Julia M. Hum
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA
| | - Gregory Bix
- Departments of Neurosurgery and Neurology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - Alexander G. Robling
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Rajesh Sardar
- Department of Chemistry and Chemical Biology, School of Science, Indiana University, Indianapolis, IN 46202, USA
| | - Mary C. Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - William R. Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA
- Correspondence:
| |
Collapse
|
18
|
Wang JS, Wein MN. Pathways Controlling Formation and Maintenance of the Osteocyte Dendrite Network. Curr Osteoporos Rep 2022; 20:493-504. [PMID: 36087214 PMCID: PMC9718876 DOI: 10.1007/s11914-022-00753-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the molecular mechanisms involved in osteocyte dendrite formation, summarize the similarities between osteocytic and neuronal projections, and highlight the importance of osteocyte dendrite maintenance in human skeletal disease. RECENT FINDINGS It is suggested that there is a causal relationship between the loss of osteocyte dendrites and the increased osteocyte apoptosis during conditions including aging, microdamage, and skeletal disease. A few mechanisms are proposed to control dendrite formation and outgrowth, such as via the regulation of actin polymerization dynamics. This review addresses the impact of osteocyte dendrites in bone health and disease. Recent advances in multi-omics, in vivo and in vitro models, and microscopy-based imaging have provided novel approaches to reveal the underlying mechanisms that regulate dendrite development. Future therapeutic approaches are needed to target the process of osteocyte dendrite formation.
Collapse
Affiliation(s)
- Jialiang S Wang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
19
|
Xu W, Zhu J, Hu J, Xiao L. Engineering the biomechanical microenvironment of chondrocytes towards articular cartilage tissue engineering. Life Sci 2022; 309:121043. [DOI: 10.1016/j.lfs.2022.121043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/24/2022] [Accepted: 10/02/2022] [Indexed: 11/28/2022]
|
20
|
Wang H, Du T, Li R, Main RP, Yang H. Interactive effects of various loading parameters on the fluid dynamics within the lacunar-canalicular system for a single osteocyte. Bone 2022; 158:116367. [PMID: 35181573 DOI: 10.1016/j.bone.2022.116367] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 12/26/2022]
Abstract
The osteocyte lacunar-canalicular system (LCS) serves as a mechanotransductive core where external loading applied to the skeleton is transduced into mechanical signals (e.g., fluid shear) that can be sensed by mechanosensors (osteocytes). The fluid velocity and shear stress within the LCS are affected by various loading parameters. However, the interactive effect of distinct loading parameters on the velocity and shear stress in the LCS remains unclear. To address this issue, we developed a multiscale modeling approach, combining a poroelastic finite element (FE) model with a single osteocytic LCS unit model to calculate the flow velocity and shear stress within the LCS. Next, a sensitivity analysis was performed to investigate individual and interactive effects of strain magnitude, strain rate, number of cycles, and intervening short rests between loading cycles on the velocity and shear stress around the osteocyte. Lastly, we developed a relatively simple regression model to predict those outcomes. Our results demonstrated that the strain magnitude or rate alone were the main factors affecting the velocity and shear stress; however, the combination of these two was not directly additive, and addition of a short rest between cycles could enhance the combination of these two related factors. These results show highly interactive effects of distinct loading parameters on fluid velocity and shear stress in the LCS. Specifically, our results suggest that an enhanced fluid dynamics environment in the LCS can be achieved with a brief number of load cycles combined with short rest insertion and high strain magnitude and rate.
Collapse
Affiliation(s)
- Huiru Wang
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Tianming Du
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Rui Li
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Russell P Main
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, IN, USA; Weldon School of Biomedical Engineering, Purdue University, IN, USA
| | - Haisheng Yang
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
21
|
Abstract
Disuse osteoporosis describes a state of bone loss due to local skeletal unloading or systemic immobilization. This review will discuss advances in the field that have shed light on clinical observations, mechanistic insights and options for the treatment of disuse osteoporosis. Clinical settings of disuse osteoporosis include spinal cord injury, other neurological and neuromuscular disorders, immobilization after fractures and bed rest (real or modeled). Furthermore, spaceflight-induced bone loss represents a well-known adaptive process to microgravity. Clinical studies have outlined that immobilization leads to immediate bone loss in both the trabecular and cortical compartments accompanied by relatively increased bone resorption and decreased bone formation. The fact that the low bone formation state has been linked to high levels of the osteocyte-secreted protein sclerostin is one of the many findings that has brought matrix-embedded, mechanosensitive osteocytes into focus in the search for mechanistic principles. Previous basic research has primarily involved rodent models based on tail suspension, spaceflight and other immobilization methods, which have underlined the importance of osteocytes in the pathogenesis of disuse osteoporosis. Furthermore, molecular-based in vitro and in vivo approaches have revealed that osteocytes sense mechanical loading through mechanosensors that translate extracellular mechanical signals to intracellular biochemical signals and regulate gene expression. Osteocytic mechanosensors include the osteocyte cytoskeleton and dendritic processes within the lacuno-canalicular system (LCS), ion channels (e.g., Piezo1), extracellular matrix, primary cilia, focal adhesions (integrin-based) and hemichannels and gap junctions (connexin-based). Overall, disuse represents one of the major factors contributing to immediate bone loss and osteoporosis, and alterations in osteocytic pathways appear crucial to the bone loss associated with unloading.
Collapse
Affiliation(s)
- Tim Rolvien
- Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestrasse 59, 22529, Hamburg, Germany.
| |
Collapse
|
22
|
Hayes AJ, Farrugia BL, Biose IJ, Bix GJ, Melrose J. Perlecan, A Multi-Functional, Cell-Instructive, Matrix-Stabilizing Proteoglycan With Roles in Tissue Development Has Relevance to Connective Tissue Repair and Regeneration. Front Cell Dev Biol 2022; 10:856261. [PMID: 35433700 PMCID: PMC9010944 DOI: 10.3389/fcell.2022.856261] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/28/2022] [Indexed: 12/19/2022] Open
Abstract
This review highlights the multifunctional properties of perlecan (HSPG2) and its potential roles in repair biology. Perlecan is ubiquitous, occurring in vascular, cartilaginous, adipose, lymphoreticular, bone and bone marrow stroma and in neural tissues. Perlecan has roles in angiogenesis, tissue development and extracellular matrix stabilization in mature weight bearing and tensional tissues. Perlecan contributes to mechanosensory properties in cartilage through pericellular interactions with fibrillin-1, type IV, V, VI and XI collagen and elastin. Perlecan domain I - FGF, PDGF, VEGF and BMP interactions promote embryonic cellular proliferation, differentiation, and tissue development. Perlecan domain II, an LDLR-like domain interacts with lipids, Wnt and Hedgehog morphogens. Perlecan domain III binds FGF-7 and 18 and has roles in the secretion of perlecan. Perlecan domain IV, an immunoglobulin repeat domain, has cell attachment and matrix stabilizing properties. Perlecan domain V promotes tissue repair through interactions with VEGF, VEGF-R2 and α2β1 integrin. Perlecan domain-V LG1-LG2 and LG3 fragments antagonize these interactions. Perlecan domain V promotes reconstitution of the blood brain barrier damaged by ischemic stroke and is neurogenic and neuroprotective. Perlecan-VEGF-VEGFR2, perlecan-FGF-2 and perlecan-PDGF interactions promote angiogenesis and wound healing. Perlecan domain I, III and V interactions with platelet factor-4 and megakaryocyte and platelet inhibitory receptor promote adhesion of cells to implants and scaffolds in vascular repair. Perlecan localizes acetylcholinesterase in the neuromuscular junction and is of functional significance in neuromuscular control. Perlecan mutation leads to Schwartz-Jampel Syndrome, functional impairment of the biomechanical properties of the intervertebral disc, variable levels of chondroplasia and myotonia. A greater understanding of the functional working of the neuromuscular junction may be insightful in therapeutic approaches in the treatment of neuromuscular disorders. Tissue engineering of salivary glands has been undertaken using bioactive peptides (TWSKV) derived from perlecan domain IV. Perlecan TWSKV peptide induces differentiation of salivary gland cells into self-assembling acini-like structures that express salivary gland biomarkers and secrete α-amylase. Perlecan also promotes chondroprogenitor stem cell maturation and development of pluripotent migratory stem cell lineages, which participate in diarthrodial joint formation, and early cartilage development. Recent studies have also shown that perlecan is prominently expressed during repair of adult human articular cartilage. Perlecan also has roles in endochondral ossification and bone development. Perlecan domain I hydrogels been used in tissue engineering to establish heparin binding growth factor gradients that promote cell migration and cartilage repair. Perlecan domain I collagen I fibril scaffolds have also been used as an FGF-2 delivery system for tissue repair. With the availability of recombinant perlecan domains, the development of other tissue repair strategies should emerge in the near future. Perlecan co-localization with vascular elastin in the intima, acts as a blood shear-flow endothelial sensor that regulates blood volume and pressure and has a similar role to perlecan in canalicular fluid, regulating bone development and remodeling. This complements perlecan's roles in growth plate cartilage and in endochondral ossification to form the appendicular and axial skeleton. Perlecan is thus a ubiquitous, multifunctional, and pleomorphic molecule of considerable biological importance. A greater understanding of its diverse biological roles and functional repertoires during tissue development, growth and disease will yield valuable insights into how this impressive proteoglycan could be utilized successfully in repair biology.
Collapse
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Wales, United Kingdom
| | - Brooke L. Farrugia
- Department of Biomedical Engineering, Melbourne School of Engineering, The University of Melbourne, Melbourne, VIC, Australia
| | - Ifechukwude J. Biose
- Departments of Neurosurgery and Neurology, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
| | - Gregory J. Bix
- Departments of Neurosurgery and Neurology, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital, The Faculty of Medicine and Health, The University of Sydney, St. Leonard’s, NSW, Australia
| |
Collapse
|
23
|
Choi JUA, Kijas AW, Lauko J, Rowan AE. The Mechanosensory Role of Osteocytes and Implications for Bone Health and Disease States. Front Cell Dev Biol 2022; 9:770143. [PMID: 35265628 PMCID: PMC8900535 DOI: 10.3389/fcell.2021.770143] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Bone homeostasis is a dynamic equilibrium between bone-forming osteoblasts and bone-resorbing osteoclasts. This process is primarily controlled by the most abundant and mechanosensitive bone cells, osteocytes, that reside individually, within chambers of porous hydroxyapatite bone matrix. Recent studies have unveiled additional functional roles for osteocytes in directly contributing to local matrix regulation as well as systemic roles through endocrine functions by communicating with distant organs such as the kidney. Osteocyte function is governed largely by both biochemical signaling and the mechanical stimuli exerted on bone. Mechanical stimulation is required to maintain bone health whilst aging and reduced level of loading are known to result in bone loss. To date, both in vivo and in vitro approaches have been established to answer important questions such as the effect of mechanical stimuli, the mechanosensors involved, and the mechanosensitive signaling pathways in osteocytes. However, our understanding of osteocyte mechanotransduction has been limited due to the technical challenges of working with these cells since they are individually embedded within the hard hydroxyapatite bone matrix. This review highlights the current knowledge of the osteocyte functional role in maintaining bone health and the key regulatory pathways of these mechanosensitive cells. Finally, we elaborate on the current therapeutic opportunities offered by existing treatments and the potential for targeting osteocyte-directed signaling.
Collapse
Affiliation(s)
- Jung Un Ally Choi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Amanda W Kijas
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Jan Lauko
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Alan E Rowan
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
24
|
Kahle ER, Han B, Chandrasekaran P, Phillips ER, Mulcahey MK, Lu XL, Marcolongo MS, Han L. Molecular Engineering of Pericellular Microniche via Biomimetic Proteoglycans Modulates Cell Mechanobiology. ACS NANO 2022; 16:1220-1230. [PMID: 35015500 PMCID: PMC9271520 DOI: 10.1021/acsnano.1c09015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Molecular engineering of biological tissues using synthetic mimics of native matrix molecules can modulate the mechanical properties of the cellular microenvironment through physical interactions with existing matrix molecules, and in turn, mediate the corresponding cell mechanobiology. In articular cartilage, the pericellular matrix (PCM) is the immediate microniche that regulates cell fate, signaling, and metabolism. The negatively charged osmo-environment, as endowed by PCM proteoglycans, is a key biophysical cue for cell mechanosensing. This study demonstrated that biomimetic proteoglycans (BPGs), which mimic the ultrastructure and polyanionic nature of native proteoglycans, can be used to molecularly engineer PCM micromechanics and cell mechanotransduction in cartilage. Upon infiltration into bovine cartilage explant, we showed that localization of BPGs in the PCM leads to increased PCM micromodulus and enhanced chondrocyte intracellular calcium signaling. Applying molecular force spectroscopy, we revealed that BPGs integrate with native PCM through augmenting the molecular adhesion of aggrecan, the major PCM proteoglycan, at the nanoscale. These interactions are enabled by the biomimetic "bottle-brush" ultrastructure of BPGs and facilitate the integration of BPGs within the PCM. Thus, this class of biomimetic molecules can be used for modulating molecular interactions of pericellular proteoglycans and harnessing cell mechanosensing. Because the PCM is a prevalent feature of various cell types, BPGs hold promising potential for improving regeneration and disease modification for not only cartilage-related healthcare but many other tissues and diseases.
Collapse
Affiliation(s)
- Elizabeth R. Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Evan R. Phillips
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
| | - Mary K. Mulcahey
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Michele S. Marcolongo
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
- Department of Mechanical Engineering, Villanova University, Villanova, PA 19085, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
25
|
Abstract
Fracture healing is a complex, multistep process that is highly sensitive to mechanical signaling. To optimize repair, surgeons prescribe immediate weight-bearing as-tolerated within 24 hours after surgical fixation; however, this recommendation is based on anecdotal evidence and assessment of bulk healing outcomes (e.g., callus size, bone volume, etc.). Given challenges in accurately characterizing the mechanical environment and the ever-changing properties of the regenerate, the principles governing mechanical regulation of repair, including their cell and molecular basis, are not yet well defined. However, the use of mechanobiological rodent models, and their relatively large genetic toolbox, combined with recent advances in imaging approaches and single-cell analyses is improving our understanding of the bone microenvironment in response to loading. This review describes the identification and characterization of distinct cell populations involved in bone healing and highlights the most recent findings on mechanical regulation of bone homeostasis and repair with an emphasis on osteo-angio coupling. A discussion on aging and its impact on bone mechanoresponsiveness emphasizes the need for novel mechanotherapeutics that can re-sensitize skeletal stem and progenitor cells to physical rehabilitation protocols.
Collapse
Affiliation(s)
- Tareq Anani
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA
| | - Alesha B Castillo
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA; Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 11201, USA; Department of Veterans Affairs, New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010, USA.
| |
Collapse
|
26
|
Osteocytic Pericellular Matrix (PCM): Accelerated Degradation under In Vivo Loading and Unloading Conditions Using a Novel Imaging Approach. Genes (Basel) 2021; 13:genes13010072. [PMID: 35052411 PMCID: PMC8775093 DOI: 10.3390/genes13010072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 01/03/2023] Open
Abstract
The proteoglycan-containing pericellular matrix (PCM) controls both the biophysical and biochemical microenvironment of osteocytes, which are the most abundant cells embedded and dispersed in bones. As a molecular sieve, osteocytic PCMs not only regulate mass transport to and from osteocytes but also act as sensors of external mechanical environments. The turnover of osteocytic PCM remains largely unknown due to technical challenges. Here, we report a novel imaging technique based on metabolic labeling and “click-chemistry,” which labels de novo PCM as “halos” surrounding osteocytes in vitro and in vivo. We then tested the method and showed different labeling patterns in young vs. old bones. Further “pulse-chase” experiments revealed dramatic difference in the “half-life” of PCM of cultured osteocytes (~70 h) and that of osteocytes in vivo (~75 d). When mice were subjected to either 3-week hindlimb unloading or 7-week tibial loading (5.1 N, 4 Hz, 3 d/week), PCM half-life was shortened (~20 d) and degradation accelerated. Matrix metallopeptidase MMP-14 was elevated in mechanically loaded osteocytes, which may contribute to PCM degradation. This study provides a detailed procedure that enables semi-quantitative study of the osteocytic PCM remodeling in vivo and in vitro.
Collapse
|
27
|
Kameo Y, Ozasa M, Adachi T. Computational framework for analyzing flow-induced strain on osteocyte as modulated by microenvironment. J Mech Behav Biomed Mater 2021; 126:105027. [PMID: 34920322 DOI: 10.1016/j.jmbbm.2021.105027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 12/02/2021] [Indexed: 12/26/2022]
Abstract
Osteocytes buried in bone matrix are major mechanosensory cells that regulate bone remodeling in response to interstitial fluid flow in a lacuno-canalicular porosity. To gain an understanding of the mechanism of osteocyte mechanosensing, it is important to be able to evaluate the local strain on the osteocyte process membrane induced by the interstitial fluid flow. The microenvironment of the osteocytes, including the pericellular matrix (PCM) and canalicular ultrastructure, is a key modulator of the flow-induced strain on the osteocyte process membrane because it produces heterogeneous flow patterns in the pericellular space. To investigate the effect of changes in the microenvironment of osteocytes on the flow-induced strain, we developed a novel computational framework for analyzing the fluid-structure interaction. Computer simulations based on the proposed framework enabled evaluation of the spatial distribution of flow-induced strain on the osteocyte process membrane according to changes in the PCM density and canalicular curvature. The simulation results reveal that a decrease in PCM density and an increase in canalicular curvature, each of which is associated with aging and bone disease, have the notable effect of enhancing local flow-induced strain on the osteocyte process membrane. We believe that the proposed computational framework is a promising framework for investigating cell-specific mechanical stimuli and that it has the potential to accelerate the mechanobiological study of osteocytes by providing a deeper understanding of their mechanical environment in living bone tissue.
Collapse
Affiliation(s)
- Yoshitaka Kameo
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masahiro Ozasa
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taiji Adachi
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Postmenopausal osteoporosis reduces circulating estrogen levels, which leads to osteoclast resorption, bone loss, and fracture. This review addresses emerging evidence that osteoporosis is not simply a disease of bone loss but that mechanosensitive osteocytes that regulate both osteoclasts and osteoblasts are also impacted by estrogen deficiency. RECENT FINDINGS At the onset of estrogen deficiency, the osteocyte mechanical environment is altered, which coincides with temporal changes in bone tissue composition. The osteocyte microenvironment is also altered, apoptosis is more prevalent, and hypermineralization occurs. The mechanobiological responses of osteocytes are impaired under estrogen deficiency, which exacerbates osteocyte paracrine regulation of osteoclasts. Recent research reveals changes in osteocytes during estrogen deficiency that may play a critical role in the etiology of the disease. A paradigm change for osteoporosis therapy requires an advanced understanding of such changes to establish the efficacy of osteocyte-targeted therapies to inhibit resorption and secondary mineralization.
Collapse
Affiliation(s)
- Laoise M McNamara
- Mechanobiology and Medical Device Research Group, Biomedical Engineering, College of Science and Engineering, National University of Ireland, Galway, Ireland.
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland.
| |
Collapse
|
29
|
Wang S, Pei S, Wasi M, Parajuli A, Yee A, You L, Wang L. Moderate tibial loading and treadmill running, but not overloading, protect adult murine bone from destruction by metastasized breast cancer. Bone 2021; 153:116100. [PMID: 34246808 PMCID: PMC8478818 DOI: 10.1016/j.bone.2021.116100] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Osteolytic bone lesions, which develop in many metastatic breast cancer patients, impair bone integrity and lead to adverse skeletal related events that are difficult to treat and sometimes fatal. Moderate mechanical loading has been shown to suppress osteolysis in young mice with breast cancer. In this study, we aimed to investigate the dose-dependent effects of mechanical loading on protecting the integrity of adult skeletons with breast cancer. Localized tibial loading and aerobic treadmill running with three levels of varying intensity were tested in a syngeneic mammary tumor bone metastasis model. Adult C57BL/6J female mice (14-week-old, N = 88 mice) received intra-tibial injections of Py8119 triple-negative murine breast cancer cells or PBS and underwent 4 to 5 weeks of exercise or acted as sedentary/non-loaded controls. The bone structure was monitored longitudinally with weekly in vivo micro-computed tomography imaging, while the cellular responses in bone and marrow were examined using immunohistochemistry. Moderate treadmill running (16 m/min, 50 min/day, 5 days/week, and 5 weeks) and tibial loading (4.5 N, 630 με, 4 Hz, 300 cycles/day, 5 days/week, and 4 weeks) suppressed tumor-induced bone destruction, as evaluated by full-thickness perforation of tibial cortex and the volume of osteolytic lesions in the cortex. In contrast, tibial loading at higher magnitude (8 N, 1100 με) induced woven bone and accelerated bone destruction, compared with the non-loaded controls. The three exercise regimens differentially affected osteocyte apoptosis, osteocyte hypoxia, osteoclast activity, bone marrow vasculature, and tumor proliferation. In conclusion, the relationship between exercise intensity and the risk of breast cancer-induced osteolysis was found to follow a J-shaped curve in a preclinical model, suggesting the need to optimize exercise parameters in order to harness the skeletal benefits of exercise in metastatic breast cancers.
Collapse
Affiliation(s)
- Shubo Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Shaopeng Pei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Murtaza Wasi
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Ashutosh Parajuli
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Albert Yee
- Division of Orthopaedics, Department of Surgery, Sunnybrook Health Sciences Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
30
|
Xu Z, Chen S, Feng D, Liu Y, Wang Q, Gao T, Liu Z, Zhang Y, Chen J, Qiu L. Biological role of heparan sulfate in osteogenesis: A review. Carbohydr Polym 2021; 272:118490. [PMID: 34420746 DOI: 10.1016/j.carbpol.2021.118490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022]
Abstract
Heparan sulfate (HS) is extensively expressed in cells, for example, cell membrane and extracellular matrix of most mammalian cells and tissues, playing a key role in the growth and development of life by maintaining homeostasis and implicating in the etiology and diseases. Recent studies have revealed that HS is involved in osteogenesis via coordinating multiple signaling pathways. The potential effect of HS on osteogenesis is a complicated and delicate biological process, which involves the participation of osteocytes, chondrocytes, osteoblasts, osteoclasts and a variety of cytokines. In this review, we summarized the structural and functional characteristics of HS and highlighted the molecular mechanism of HS in bone metabolism to provide novel research perspectives for the further medical research.
Collapse
Affiliation(s)
- Zhujie Xu
- Department of Orthopedics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Shayang Chen
- Department of Orthopedics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Dehong Feng
- Department of Orthopedics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Yi Liu
- Department of Orthopedics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China.
| | - Qiqi Wang
- Department of Orthopedics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Tianshu Gao
- Department of Orthopedics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Zhenwei Liu
- Department of Orthopedics, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Yan Zhang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Jinghua Chen
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Lipeng Qiu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
31
|
Lai X, Chung R, Li Y, Liu XS, Wang L. Lactation alters fluid flow and solute transport in maternal skeleton: A multiscale modeling study on the effects of microstructural changes and loading frequency. Bone 2021; 151:116033. [PMID: 34102350 PMCID: PMC8276854 DOI: 10.1016/j.bone.2021.116033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/23/2021] [Accepted: 06/01/2021] [Indexed: 01/02/2023]
Abstract
The female skeleton undergoes significant material and ultrastructural changes to meet high calcium demands during reproduction and lactation. Through the peri-lacunar/canalicular remodeling (PLR), osteocytes actively resorb surrounding matrix and enlarge their lacunae and canaliculi during lactation, which are quickly reversed after weaning. How these changes alter the physicochemical environment of osteocytes, the most abundant and primary mechanosensing cells in bone, are not well understood. In this study, we developed a multiscale poroelastic modeling technique to investigate lactation-induced changes in stress, fluid pressurization, fluid flow, and solute transport across multiple length scales (whole bone, porous midshaft cortex, lacunar-canalicular pore system (LCS), and pericellular matrix (PCM) around osteocytes) in murine tibiae subjected to axial compression at 3 N peak load (~320 με) at 0.5, 2, or 4 Hz. Based on previously reported skeletal anatomical measurements from lactating and nulliparous mice, our models demonstrated that loading frequency, LCS porosity, and PCM density were major determinants of fluid and solute flows responsible for osteocyte mechanosensing, cell-cell signaling, and metabolism. When loaded at 0.5 Hz, lactation-induced LCS expansion and potential PCM reduction promoted solute transport and osteocyte mechanosensing via primary cilia, but suppressed mechanosensing via fluid shear and/or drag force on the cell membrane. Interestingly, loading at 2 or 4 Hz was found to overcome the mechanosensing deficits observed at 0.5 Hz and these counter effects became more pronounced at 4 Hz and with sparser PCM in the lactating bone. Synergistically, higher loading frequency (2, 4 Hz) and sparser PCM enhanced flow-mediated mechanosensing and diffusion/convection of nutrients and signaling molecules for osteocytes. In summary, lactation-induced structural changes alter the local environment of osteocytes in ways that favor metabolism, mechanosensing, and post-weaning recovery of maternal bone. Thus, osteocytes play a role in balancing the metabolic and mechanical functions of female skeleton during reproduction and lactation.
Collapse
Affiliation(s)
- Xiaohan Lai
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Rebecca Chung
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yihan Li
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiaowei Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, United States.
| |
Collapse
|
32
|
Li Y, de Bakker CMJ, Lai X, Zhao H, Parajuli A, Tseng WJ, Pei S, Meng T, Chung R, Wang L, Liu XS. Maternal bone adaptation to mechanical loading during pregnancy, lactation, and post-weaning recovery. Bone 2021; 151:116031. [PMID: 34098162 PMCID: PMC8504362 DOI: 10.1016/j.bone.2021.116031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/25/2021] [Accepted: 06/01/2021] [Indexed: 12/16/2022]
Abstract
The maternal skeleton undergoes dramatic bone loss during pregnancy and lactation, and substantial bone recovery post-weaning. The structural adaptations of maternal bone during reproduction and lactation exert a better protection of the mechanical integrity at the critical load-bearing sites, suggesting the importance of physiological load-bearing in regulating reproduction-induced skeletal alterations. Although it is suggested that physical exercise during pregnancy and breastfeeding improves women's physical and psychological well-being, its effects on maternal bone health remain unclear. Therefore, the objective of this study was to investigate the maternal bone adaptations to external mechanical loading during pregnancy, lactation, and post-weaning recovery. By utilizing an in vivo dynamic tibial loading protocol in a rat model, we demonstrated improved maternal cortical bone structure in response to dynamic loading at tibial midshaft, regardless of reproductive status. Notably, despite the minimal loading responses detected in the trabecular bone in virgins, rat bone during lactation experienced enhanced mechano-responsiveness in both trabecular and cortical bone compartments when compared to rats at other reproductive stages or age-matched virgins. Furthermore, our study showed that the lactation-induced elevation in osteocyte peri-lacunar/canalicular remodeling (PLR) activities led to enlarged osteocyte lacunae. This may result in alterations in interstitial fluid flow-mediated mechanical stimulation on osteocytes and an elevation in solute transport through the lacunar-canalicular system (LCS) during high-frequency dynamic loading, thus enhancing mechano-responsiveness of maternal bone during lactation. Taken together, findings from this study provide important insights into the relationship between reproduction- and lactation-induced skeletal changes and external mechanical loading, emphasizing the importance of weight-bearing exercise on maternal bone health during reproduction and postpartum.
Collapse
Affiliation(s)
- Yihan Li
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Chantal M J de Bakker
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Radiology, Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - Xiaohan Lai
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongbo Zhao
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ashutosh Parajuli
- Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Wei-Ju Tseng
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Shaopeng Pei
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Tan Meng
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rebecca Chung
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liyun Wang
- Center for Biomechanical Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - X Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
33
|
Kim J, Kigami H, Adachi T. Comparative gene expression analysis for pre-osteoblast MC3T3-E1 cells under non-adhesive culture toward osteocyte differentiation. J Biosci Bioeng 2021; 132:651-656. [PMID: 34556421 DOI: 10.1016/j.jbiosc.2021.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/26/2022]
Abstract
Osteocytes play an important role to modulate the bone remodeling and are also known as terminally differentiated cells originated from the osteoblast precursor cells, but its differentiation mechanism remains unclear. Since an efficient in vitro method to evoke the osteocyte differentiation from the osteoblast precursor cells has not been established, we conducted the comparative gene expression analysis for mouse pre-osteoblast MC3T3-E1 cells in order to elucidate the key factors to induce the osteocyte differentiation from the pre-osteoblast cells. In this study, we prepared four different culture environments by modulating their cell-substrate interaction and cell-cell interaction; (i) low and (ii) high cell density on the adhesive culture models, and (iii) low and (iv) high cell density on the non-adhesive floating culture models. By comparing these conditions in terms of cell-substrate and cell-cell interaction, we showed that the elimination of cell-substrate interaction under non-adhesive floating culture greatly up-regulated the gene expression of osteocyte markers in the pre-osteoblast cells. Moreover, the presence of moderate cell-cell interaction in the non-adhesive spheroid culture further enhanced the up-regulation of osteocyte markers for the pre-osteoblast cells. The results altogether suggest the most appropriate culture environment to induce the in vitro osteocyte differentiation of pre-osteoblast cells.
Collapse
Affiliation(s)
- Jeonghyun Kim
- Department of Mechanical Systems Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Hiroyuki Kigami
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto 606-8507, Japan
| | - Taiji Adachi
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto 606-8507, Japan; Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
34
|
Yamamoto T, Hasegawa T, Fraitas PHLD, Hongo H, Zhao S, Yamamoto T, Nasoori A, Abe M, Maruoka H, Kubota K, Morimoto Y, Haraguchi M, Shimizu T, Takahata M, Iwasaki N, Li M, Amizuka N. Histochemical characteristics on minimodeling-based bone formation induced by anabolic drugs for osteoporotic treatment. Biomed Res 2021; 42:161-171. [PMID: 34544992 DOI: 10.2220/biomedres.42.161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Modeling, the changes of bone size and shape, often takes place at the developmental stages, whereas bone remodeling-replacing old bone with new bone-predominantly occurs in adults. Unlike bone remodeling, bone formation induced by modeling i.e., minimodeling (microscopic modeling in cancellous bone) is independent of osteoclastic bone resorption. Although recently-developed drugs for osteoporotic treatment could induce minimodeling-based bone formation in addition to remodeling-based bone formation, few reports have demonstrated the histological aspects of minimodeling-based bone formation. After administration of eldecalcitol or romosozumab, unlike teriparatide treatment, mature osteoblasts formed new bone by minimodeling, without developing thick preosteoblastic layers. The histological characteristics of minimodeling-based bone formation is quite different from remodeling, as it is not related to osteoclastic bone resorption, resulting in convex-shaped new bone and smooth cement lines called arrest lines. In this review, we will show histological properties of minimodeling-based bone formation by osteoporotic drugs.
Collapse
Affiliation(s)
- Tomomaya Yamamoto
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University.,Northern Army Medical Unit, Camp Makomanai, Japan Ground Self-Defense Forces
| | - Tomoka Hasegawa
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | | | - Hiromi Hongo
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Shen Zhao
- National Clinical Research Center of Stomatology, Department of Endodontics, School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Ninth People's Hospital, Shanghai Jiaotong University
| | - Tsuneyuki Yamamoto
- Oral Functional Anatomy, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Alireza Nasoori
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Miki Abe
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Haruhi Maruoka
- Orthodontics, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Keisuke Kubota
- Oral Functional Prosthodontics, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Yasuhito Morimoto
- Periodontology and Endodontology, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Mai Haraguchi
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Tomohiro Shimizu
- Department of Orthopedic Surgery, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Masahiko Takahata
- Department of Orthopedic Surgery, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| | - Minqi Li
- Shandong Provincial Key Laboratory of Oral Biomedicine, The School of Stomatology, Shandong University
| | - Norio Amizuka
- Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, and Faculty of Dental Medicine, Hokkaido University
| |
Collapse
|
35
|
Yokoyama Y, Kameo Y, Kamioka H, Adachi T. High-resolution image-based simulation reveals membrane strain concentration on osteocyte processes caused by tethering elements. Biomech Model Mechanobiol 2021; 20:2353-2360. [PMID: 34471950 PMCID: PMC8595188 DOI: 10.1007/s10237-021-01511-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/13/2021] [Indexed: 11/08/2022]
Abstract
Osteocytes are vital for regulating bone remodeling by sensing the flow-induced mechanical stimuli applied to their cell processes. In this mechanosensing mechanism, tethering elements (TEs) connecting the osteocyte process with the canalicular wall potentially amplify the strain on the osteocyte processes. The ultrastructure of the osteocyte processes and canaliculi can be visualized at a nanometer scale using high-resolution imaging via ultra-high voltage electron microscopy (UHVEM). Moreover, the irregular shapes of the osteocyte processes and the canaliculi, including the TEs in the canalicular space, should considerably influence the mechanical stimuli applied to the osteocytes. This study aims to characterize the roles of the ultrastructure of osteocyte processes and canaliculi in the mechanism of osteocyte mechanosensing. Thus, we constructed a high-resolution image-based model of an osteocyte process and a canaliculus using UHVEM tomography and investigated the distribution and magnitude of flow-induced local strain on the osteocyte process by performing fluid–structure interaction simulation. The analysis results reveal that local strain concentration in the osteocyte process was induced by a small number of TEs with high tension, which were inclined depending on the irregular shapes of osteocyte processes and canaliculi. Therefore, this study could provide meaningful insights into the effect of ultrastructure of osteocyte processes and canaliculi on the osteocyte mechanosensing mechanism.
Collapse
Affiliation(s)
- Yuka Yokoyama
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshitaka Kameo
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-ku, Okayama, 700-8525, Japan
| | - Taiji Adachi
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
36
|
Hagan ML, Balayan V, McGee-Lawrence ME. Plasma membrane disruption (PMD) formation and repair in mechanosensitive tissues. Bone 2021; 149:115970. [PMID: 33892174 PMCID: PMC8217198 DOI: 10.1016/j.bone.2021.115970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/26/2021] [Accepted: 04/17/2021] [Indexed: 01/04/2023]
Abstract
Mammalian cells employ an array of biological mechanisms to detect and respond to mechanical loading in their environment. One such mechanism is the formation of plasma membrane disruptions (PMD), which foster a molecular flux across cell membranes that promotes tissue adaptation. Repair of PMD through an orchestrated activity of molecular machinery is critical for cell survival, and the rate of PMD repair can affect downstream cellular signaling. PMD have been observed to influence the mechanical behavior of skin, alveolar, and gut epithelial cells, aortic endothelial cells, corneal keratocytes and epithelial cells, cardiac and skeletal muscle myocytes, neurons, and most recently, bone cells including osteoblasts, periodontal ligament cells, and osteocytes. PMD are therefore positioned to affect the physiological behavior of a wide range of vertebrate organ systems including skeletal and cardiac muscle, skin, eyes, the gastrointestinal tract, the vasculature, the respiratory system, and the skeleton. The purpose of this review is to describe the processes of PMD formation and repair across these mechanosensitive tissues, with a particular emphasis on comparing and contrasting repair mechanisms and downstream signaling to better understand the role of PMD in skeletal mechanobiology. The implications of PMD-related mechanisms for disease and potential therapeutic applications are also explored.
Collapse
Affiliation(s)
- Mackenzie L Hagan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA
| | - Vanshika Balayan
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA; Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
37
|
What Are the Potential Roles of Nuclear Perlecan and Other Heparan Sulphate Proteoglycans in the Normal and Malignant Phenotype. Int J Mol Sci 2021; 22:ijms22094415. [PMID: 33922532 PMCID: PMC8122901 DOI: 10.3390/ijms22094415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/27/2022] Open
Abstract
The recent discovery of nuclear and perinuclear perlecan in annulus fibrosus and nucleus pulposus cells and its known matrix stabilizing properties in tissues introduces the possibility that perlecan may also have intracellular stabilizing or regulatory roles through interactions with nuclear envelope or cytoskeletal proteins or roles in nucleosomal-chromatin organization that may regulate transcriptional factors and modulate gene expression. The nucleus is a mechano-sensor organelle, and sophisticated dynamic mechanoresponsive cytoskeletal and nuclear envelope components support and protect the nucleus, allowing it to perceive and respond to mechano-stimulation. This review speculates on the potential roles of perlecan in the nucleus based on what is already known about nuclear heparan sulphate proteoglycans. Perlecan is frequently found in the nuclei of tumour cells; however, its specific role in these diseased tissues is largely unknown. The aim of this review is to highlight probable roles for this intriguing interactive regulatory proteoglycan in the nucleus of normal and malignant cell types.
Collapse
|
38
|
Guilak F, Hayes AJ, Melrose J. Perlecan in Pericellular Mechanosensory Cell-Matrix Communication, Extracellular Matrix Stabilisation and Mechanoregulation of Load-Bearing Connective Tissues. Int J Mol Sci 2021; 22:2716. [PMID: 33800241 PMCID: PMC7962540 DOI: 10.3390/ijms22052716] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
In this study, we review mechanoregulatory roles for perlecan in load-bearing connective tissues. Perlecan facilitates the co-acervation of tropoelastin and assembly of elastic microfibrils in translamellar cross-bridges which, together with fibrillin and elastin stabilise the extracellular matrix of the intervertebral disc annulus fibrosus. Pericellular perlecan interacts with collagen VI and XI to define and stabilize this matrix compartment which has a strategic position facilitating two-way cell-matrix communication between the cell and its wider extracellular matrix. Cues from the extracellular matrix are fed through this pericellular matrix back to the chondrocyte, allowing it to perceive and respond to subtle microenvironmental changes to regulate tissue homeostasis. Thus perlecan plays a key regulatory role in chondrocyte metabolism, and in chondrocyte differentiation. Perlecan acts as a transport proteoglycan carrying poorly soluble, lipid-modified proteins such as the Wnt or Hedgehog families facilitating the establishment of morphogen gradients that drive tissue morphogenesis. Cell surface perlecan on endothelial cells or osteocytes acts as a flow sensor in blood and the lacunar canalicular fluid providing feedback cues to smooth muscle cells regulating vascular tone and blood pressure, and the regulation of bone metabolism by osteocytes highlighting perlecan's multifaceted roles in load-bearing connective tissues.
Collapse
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA;
- Shriners Hospitals for Children—St. Louis, St. Louis, MO 63110, USA
| | - Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF10 3AX, UK;
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School, Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
39
|
Garcia J, McCarthy HS, Kuiper JH, Melrose J, Roberts S. Perlecan in the Natural and Cell Therapy Repair of Human Adult Articular Cartilage: Can Modifications in This Proteoglycan Be a Novel Therapeutic Approach? Biomolecules 2021; 11:biom11010092. [PMID: 33450893 PMCID: PMC7828356 DOI: 10.3390/biom11010092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/29/2022] Open
Abstract
Articular cartilage is considered to have limited regenerative capacity, which has led to the search for therapies to limit or halt the progression of its destruction. Perlecan, a multifunctional heparan sulphate (HS) proteoglycan, promotes embryonic cartilage development and stabilises the mature tissue. We investigated the immunolocalisation of perlecan and collagen between donor-matched biopsies of human articular cartilage defects (n = 10 × 2) that were repaired either naturally or using autologous cell therapy, and with age-matched normal cartilage. We explored how the removal of HS from perlecan affects human chondrocytes in vitro. Immunohistochemistry showed both a pericellular and diffuse matrix staining pattern for perlecan in both natural and cell therapy repaired cartilage, which related to whether the morphology of the newly formed tissue was hyaline cartilage or fibrocartilage. Immunostaining for perlecan was significantly greater in both these repair tissues compared to normal age-matched controls. The immunolocalisation of collagens type III and VI was also dependent on tissue morphology. Heparanase treatment of chondrocytes in vitro resulted in significantly increased proliferation, while the expression of key chondrogenic surface and genetic markers was unaffected. Perlecan was more prominent in chondrocyte clusters than in individual cells after heparanase treatment. Heparanase treatment could be a means of increasing chondrocyte responsiveness to cartilage injury and perhaps to improve repair of defects.
Collapse
Affiliation(s)
- John Garcia
- School of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, UK; (J.G.); (H.S.M.); (J.H.K.)
- Spinal Studies & Cartilage Research Group, Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK
| | - Helen S. McCarthy
- School of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, UK; (J.G.); (H.S.M.); (J.H.K.)
- Spinal Studies & Cartilage Research Group, Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK
| | - Jan Herman Kuiper
- School of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, UK; (J.G.); (H.S.M.); (J.H.K.)
- Spinal Studies & Cartilage Research Group, Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK
| | - James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Northern Sydney Area Local Health District, St. Leonards, NSW 2065, Australia;
- Sydney Medical School, Northern, The University of Sydney, Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sally Roberts
- School of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, UK; (J.G.); (H.S.M.); (J.H.K.)
- Spinal Studies & Cartilage Research Group, Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK
- Correspondence: ; Tel.: +44-1-691-404-664
| |
Collapse
|
40
|
Pei Q, Li J, Zhou P, Zhang J, Huang P, Fan J, Zou Z, Li X, Wang B. A Potential Participant in Type 2 Diabetes Bone Fragility: TIMP-1 at Sites of Osteocyte Lacunar-Canalicular System. Diabetes Metab Syndr Obes 2021; 14:4903-4909. [PMID: 34992398 PMCID: PMC8711839 DOI: 10.2147/dmso.s345081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/12/2021] [Indexed: 11/23/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with an increased risk of bone fracture, but the bone mineral density (BMD) is typically normal or higher in such patients. Because the fracture risk is independent of reduced BMD, bone fragility in T2DM may be partially due to poor bone quality. The mechanisms triggering bone quality abnormalities in T2DM are complex, and include the accumulation of advanced glycation end-products, the increased inflammation, and low bone turnover. Matrix metalloproteinases (MMPs) in bone can hydrolyze the bone matrix. Tissue inhibitors of MMPs (TIMPs) can inhibit the activity of MMPs. Both MMPs and TIMPs participate in mediating bone quality. Among all types of TIMPs, TIMP-1 is mostly reportedly increased in the serum of T2DM patients. Because osteocytes can express TIMP-1, and osteocyte pericellular matrix influences bone quality partially regulated by perilacunar/canalicular remodeling, we hypothesized that TIMP-1 at sites of osteocyte lacunar-canalicular system is involved in T2DM bone fragility.
Collapse
Affiliation(s)
- Qilin Pei
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jun Li
- Department of Orthopedic Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, 610072, People’s Republic of China
| | - Pengfei Zhou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, People’s Republic of China
| | - Jun Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Peng Huang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jingchuan Fan
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Correspondence: Bin Wang; Xi Li Email ;
| |
Collapse
|
41
|
Carina V, Della Bella E, Costa V, Bellavia D, Veronesi F, Cepollaro S, Fini M, Giavaresi G. Bone's Response to Mechanical Loading in Aging and Osteoporosis: Molecular Mechanisms. Calcif Tissue Int 2020; 107:301-318. [PMID: 32710266 DOI: 10.1007/s00223-020-00724-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Mechanotransduction is pivotal in the maintenance of homeostasis in different tissues and involves multiple cell signaling pathways. In bone, mechanical stimuli regulate the balance between bone formation and resorption; osteocytes play a central role in this regulation. Dysfunctions in mechanotransduction signaling or in osteocytes response lead to an imbalance in bone homeostasis. This alteration is very relevant in some conditions such as osteoporosis and aging. Both are characterized by increased bone weakness due to different causes, for example, the increase of osteocyte apoptosis that cause an alteration of fluid space, or the alteration of molecular pathways. There are intertwined yet very different mechanisms involved among the cell-intrinsic effects of aging on bone, the cell-intrinsic and tissue-level effects of estrogen/androgen withdrawal on bone, and the effects of reduced mechanical loading on bone, which are all involved to some degree in how aged bone fails to respond properly to stress/strain compared to younger bone. This review aims at clarifying how the cellular and molecular pathways regulated and induced in bone by mechanical stimulation are altered with aging and in osteoporosis, to highlight new possible targets for antiresorptive or anabolic bone therapeutic approaches.
Collapse
Affiliation(s)
- Valeria Carina
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy.
| | | | - Viviana Costa
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Daniele Bellavia
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Francesca Veronesi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Simona Cepollaro
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Milena Fini
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| |
Collapse
|
42
|
Melrose J. Perlecan, a modular instructive proteoglycan with diverse functional properties. Int J Biochem Cell Biol 2020; 128:105849. [PMID: 32947020 DOI: 10.1016/j.biocel.2020.105849] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/30/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022]
Abstract
This study reviewed some new aspects of the modular proteoglycan perlecan, a colossal proteoglycan with a 467 kDa core protein and five distinct functional domains. Perlecan is a heparan sulphate proteoglycan that transiently displays native CS sulphation motifs 4-C-3 and 7-D-4 during tissue morphogenesis these are expressed by progenitor cell populations during tissue development. Perlecan is susceptible to fragmentation by proteases during tissue development and in pathological tissues particularly in domains IV and V. The fragmentation pattern of domain IV has been suggested as a means of grading prostate cancer. Domain V of perlecan is of interest due to its interactive properties with integrin α5β1 that promotes pericyte migration enhancing PDGF-BB-induced phosphorylation of PDGFRβ, Src homology region 2 domain-containing phosphatase-2, and focal adhesion kinase supporting the repair of the blood brain barrier following ischaemic stroke. Fragments of domain V can also interact with α2β1 integrin disrupting tube formation by endothelial cells. LG1-LG2, LG3 fragments can antagonise VEGFR2, and α2β1 integrin interactions preventing angiogenesis by endothelial cells. These domain V fragments are of interest as potential anti-tumour agents. Perlecan attached to the luminal surfaces of endothelial cells in blood vessels acts as a flow sensor that signals back to endothelial and smooth muscle cells to regulate vascular tone and blood pressure. Perlecan also acts as a flow sensor in the lacuno-canalicular space regulating osteocytes and bone homeostasis. Along with its biomechanical regulatory properties in cartilaginous tissues this further extends the functional repertoire of this amazingly diverse functional proteoglycan.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia; Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Sydney Medical School, Northern, The University of Sydney, Australia; Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia.
| |
Collapse
|
43
|
Qin L, Liu W, Cao H, Xiao G. Molecular mechanosensors in osteocytes. Bone Res 2020; 8:23. [PMID: 32550039 PMCID: PMC7280204 DOI: 10.1038/s41413-020-0099-y] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/07/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Osteocytes, the most abundant and long-lived cells in bone, are the master regulators of bone remodeling. In addition to their functions in endocrine regulation and calcium and phosphate metabolism, osteocytes are the major responsive cells in force adaptation due to mechanical stimulation. Mechanically induced bone formation and adaptation, disuse-induced bone loss and skeletal fragility are mediated by osteocytes, which sense local mechanical cues and respond to these cues in both direct and indirect ways. The mechanotransduction process in osteocytes is a complex but exquisite regulatory process between cells and their environment, between neighboring cells, and between different functional mechanosensors in individual cells. Over the past two decades, great efforts have focused on finding various mechanosensors in osteocytes that transmit extracellular mechanical signals into osteocytes and regulate responsive gene expression. The osteocyte cytoskeleton, dendritic processes, Integrin-based focal adhesions, connexin-based intercellular junctions, primary cilium, ion channels, and extracellular matrix are the major mechanosensors in osteocytes reported so far with evidence from both in vitro and in vitro studies. This review aims to give a systematic introduction to osteocyte mechanobiology, provide details of osteocyte mechanosensors, and discuss the roles of osteocyte mechanosensitive signaling pathways in the regulation of bone homeostasis.
Collapse
Affiliation(s)
- Lei Qin
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Wen Liu
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Huiling Cao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Guozhi Xiao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| |
Collapse
|
44
|
Liu HY, Zhao S, Zhang H, Huang SY, Peng WT, Zhang CQ, Wang W. Research on solute transport behaviors in the lacunar-canalicular system using numerical simulation in microgravity. Comput Biol Med 2020; 119:103700. [PMID: 32339112 DOI: 10.1016/j.compbiomed.2020.103700] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND The lack of mass transfer in microgravity might be the underlying cause of disuse osteoporosis in astronauts after long-term space flights. The osteons are cylindrical structures and are the main structural units of the diaphysis in long bones. METHODS A multi-scale 3D fluid-solid coupled finite element model of osteon with a two-stage pore structure was developed using COMSOL software in order to investigate solute transport behaviors in the lacunar-canalicular system (LCS) induced by physiological strain loading. Certain small molecules that are necessary as solutes in tissue fluid for osteocyte metabolism were simplified to micro-particles. A comparative analysis of solute transport behaviors in the LCS induced by physiological strain loading was conducted with a frequency of 0.2-2.5 Hz in microgravity and the Earth's gravitational fields. RESULTS The average velocity of solute transport in lacunae in microgravity was 2-3 orders of magnitude lower than in Earth's gravitational field. The number of particles that represented solute transport quantity in the middle and deep lacunae increased steadily with a load frequency within the Earth's gravitational field. However, it differed based on the load frequency in microgravity, with the number of particles increasing with frequencies in the range of 0.2-0.5 Hz and 0.8-2 Hz, and decreasing with frequencies in the range of 0.5-0.8 Hz. CONCLUSIONS A moving load with appropriate frequency could promote solute transport to the middle and deep lacunae, effectively preventing apoptosis of deep osteocytes due to a lack of nutrients. The results of this study provided theoretical guidance for preventing bone loss in astronauts during long-term space flights.
Collapse
Affiliation(s)
- Hai-Ying Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, 300384, People's Republic of China.
| | - Sen Zhao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, 300384, People's Republic of China
| | - Hao Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, 300384, People's Republic of China
| | - Shuai-Yi Huang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, 300384, People's Republic of China
| | - Wan-Tao Peng
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, 300384, People's Republic of China
| | - Chun-Qiu Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, 300384, People's Republic of China
| | - Wei Wang
- Department of Mechanics, School of Mechanical Engineering, Tianjin University, Tianjin, 300354, People's Republic of China
| |
Collapse
|
45
|
Rokidi S, Bravenboer N, Gamsjaeger S, Misof B, Blouin S, Chavassieux P, Klaushofer K, Paschalis E, Papapoulos S, Appelman-Dijkstra N. Impact microindentation assesses subperiosteal bone material properties in humans. Bone 2020; 131:115110. [PMID: 31655220 DOI: 10.1016/j.bone.2019.115110] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/14/2019] [Accepted: 10/14/2019] [Indexed: 11/19/2022]
Abstract
Impact microindentation (IMI) is a Reference Point Indentation technique measuring tissue-level properties of cortical bone in humans in vivo. The nature, however, of the properties that can affect bone strength is incompletely understood. In the present study we examined bone material properties in transiliac bone biopsies obtained concurrently with measurements of Bone Material Strength index (BMSi) by IMI in 12 patients with different skeletal disorders and a wide range of BMD, with or without fractures (8 males, 4 females, mean age 48±12.2 (SD) years, range 15-60 years). IMI was performed in the mid-shaft of the right tibia with a hand-held microindenter (OsteoProbe). Cancellous and cortical bone mineralization density distributions (BMDD) were measured in the entire biopsy bone area by quantitative backscattered electron imaging. Raman measurements were obtained right at the outer edge of the cortex, and 5, 50, 100, 500μm inwards. The calculated parameters were: i) Mineral and organic matrix content as well as the mineral / matrix ratio. ii) Nanoporosity. iii) Glycosaminoglycan content. iv) Pyridinoline content. v) Maturity/crystallinity of the apatite crystallites. There was no relationship between BMSi values with any measurement of mineral content of whole bone tissue (BMD, BMDD) or maturity/crystallinity of bone mineral. On the other hand, a positive correlation between BMSi and local mineral content, and an inverse correlation between BMSi and nanoporosity at the mineralized subperiosteal edge of the sample and at 5μm inwards was found. A positive correlation was also observed between BMSi and pyridinoline content at the same locations. These results indicate that local mineral content, nanoporosity and pyridinoline content at the subperiosteal site in the transiliac bone biopsy are linked to the BMSi values measured in the tibia. As both high porosity at the nano level and low pyridinoline content of the bone matrix can negatively impact bone strength, our findings suggest that BMSi most likely assesses subperiosteal bone material properties.
Collapse
Affiliation(s)
- Stamatia Rokidi
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Viennese sickness insurance funds (WGKK) and Research funds of the Austrian workers compensation board (AUVA) Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital Vienna, Austria
| | - Natalie Bravenboer
- Leiden Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands
| | - Sonja Gamsjaeger
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Viennese sickness insurance funds (WGKK) and Research funds of the Austrian workers compensation board (AUVA) Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital Vienna, Austria
| | - Barbara Misof
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Viennese sickness insurance funds (WGKK) and Research funds of the Austrian workers compensation board (AUVA) Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital Vienna, Austria
| | - Stéphane Blouin
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Viennese sickness insurance funds (WGKK) and Research funds of the Austrian workers compensation board (AUVA) Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital Vienna, Austria
| | | | - Klaus Klaushofer
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Viennese sickness insurance funds (WGKK) and Research funds of the Austrian workers compensation board (AUVA) Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital Vienna, Austria
| | - Eleftherios Paschalis
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Viennese sickness insurance funds (WGKK) and Research funds of the Austrian workers compensation board (AUVA) Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital Vienna, Austria.
| | - Socrates Papapoulos
- Leiden Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
46
|
Main RP, Shefelbine SJ, Meakin LB, Silva MJ, van der Meulen MC, Willie BM. Murine Axial Compression Tibial Loading Model to Study Bone Mechanobiology: Implementing the Model and Reporting Results. J Orthop Res 2020; 38:233-252. [PMID: 31508836 PMCID: PMC9344861 DOI: 10.1002/jor.24466] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/23/2019] [Indexed: 02/04/2023]
Abstract
In vivo, tibial loading in mice is increasingly used to study bone adaptation and mechanotransduction. To achieve standardized and defined experimental conditions, loading parameters and animal-related factors must be considered when performing in vivo loading studies. In this review, we discuss these loading and animal-related experimental conditions, present methods to assess bone adaptation, and suggest reporting guidelines. This review originated from presentations by each of the authors at the workshop "Developing Best Practices for Mouse Models of In Vivo Loading" during the Preclinical Models Section at the Orthopaedic Research Society Annual Meeting, San Diego, CA, March 2017. Following the meeting, the authors engaged in detailed discussions with consideration of relevant literature. The guidelines and recommendations in this review are provided to help researchers perform in vivo loading experiments in mice, and thus further our knowledge of bone adaptation and the mechanisms involved in mechanotransduction. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:233-252, 2020.
Collapse
Affiliation(s)
- Russell P. Main
- Department of Basic Medical Sciences and Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA,Corresponding author: Russell Main ()
| | - Sandra J. Shefelbine
- Department of Bioengineering, Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, USA
| | - Lee B. Meakin
- Bristol Veterinary School, University of Bristol, Langford, Bristol BS40 5DU, UK
| | - Matthew J. Silva
- Departments of Orthopaedic Surgery and Biomedical Engineering, Musculoskeletal Research Center, Washington University, Saint Louis, MO, USA
| | - Marjolein C.H van der Meulen
- Meinig School of Biomedical Engineering and Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Bettina M. Willie
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| |
Collapse
|
47
|
Pei S, Parthasarathy S, Parajuli A, Martinez J, Lv M, Jiang S, Wu D, Wei S, Lu XL, Farach-Carson MC, Kirn-Safran CB, Wang L. Perlecan/Hspg2 deficiency impairs bone's calcium signaling and associated transcriptome in response to mechanical loading. Bone 2020; 131:115078. [PMID: 31715337 PMCID: PMC6945981 DOI: 10.1016/j.bone.2019.115078] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 10/25/2022]
Abstract
Perlecan, a heparan sulfate proteoglycan, acts as a mechanical sensor for bone to detect external loading. Deficiency of perlecan increases the risk of osteoporosis in patients with Schwartz-Jampel Syndrome (SJS) and attenuates loading-induced bone formation in perlecan deficient mice (Hypo). Considering that intracellular calcium [Ca2+]i is an ubiquitous messenger controlling numerous cellular processes including mechanotransduction, we hypothesized that perlecan deficiency impairs bone's calcium signaling in response to loading. To test this, we performed real-time [Ca2+]i imaging on in situ osteocytes of adult murine tibiae under cyclic loading (8N). Relative to wild type (WT), Hypo osteocytes showed decreases in the overall [Ca2+]i response rate (-58%), calcium peaks (-33%), cells with multiple peaks (-53%), peak magnitude (-6.8%), and recovery speed to baseline (-23%). RNA sequencing and pathway analysis of tibiae from mice subjected to one or seven days of unilateral loading demonstrated that perlecan deficiency significantly suppressed the calcium signaling, ECM-receptor interaction, and focal adhesion pathways following repetitive loading. Defects in the endoplasmic reticulum (ER) calcium cycling regulators such as Ryr1/ryanodine receptors and Atp2a1/Serca1 calcium pumps were identified in Hypo bones. Taken together, impaired calcium signaling may contribute to bone's reduced anabolic response to loading, underlying the osteoporosis risk for the SJS patients.
Collapse
Affiliation(s)
- Shaopeng Pei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | | | - Ashutosh Parajuli
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Jerahme Martinez
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Mengxi Lv
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Sida Jiang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center, Houston, TX 77054, United States
| | - Shuo Wei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - X Lucas Lu
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center, Houston, TX 77054, United States
| | - Catherine B Kirn-Safran
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States; Department of Biology, Widener University, Chester, PA 19013, United States
| | - Liyun Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States; Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States; Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
48
|
Parajuli A, Pei S, Zhao H, Martinez JR, Lu XL, Liu XS, Farach-Carson MC, Kirn-Safran CB, Wang L. Trabecular Bone Deficit and Enhanced Anabolic Response to Re-Ambulation after Disuse in Perlecan-Deficient Skeleton. Biomolecules 2020; 10:biom10020198. [PMID: 32013135 PMCID: PMC7072656 DOI: 10.3390/biom10020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 11/23/2022] Open
Abstract
Perlecan/Hspg2, a large monomeric heparan sulfate proteoglycan, is found in the basement membrane and extracellular matrix, where it acts as a matrix scaffold, growth factor depot, and tissue barrier. Perlecan deficiency leads to skeletal dysplasia in Schwartz-Jampel Syndrome (SJS) and is a risk factor for osteoporosis. In the SJS-mimicking murine model (Hypo), inferior cortical bone quality and impaired mechanotransduction in osteocytes were reported. This study focused on trabecular bone, where perlecan deficiency was hypothesized to result in structural deficit and altered response to disuse and re-loading. We compared the Hypo versus WT trabecular bone in both axial and appendicular skeletons of 8-38-week-old male mice, and observed severe trabecular deficit in Hypo mice, approximately 50% reduction of Tb.BV/TV regardless of skeletal site and animal age. Defects in endochondral ossification (e.g., accelerated mineralization), increases in osteoclast activity, and altered differentiation of bone progenitor cells in marrow contributed to the Hypo phenotype. The Hypo trabecular bone deteriorated further under three-week hindlimb suspension as did the WT. Re-ambulation partially recovered the lost trabecular bone in Hypo, but not in WT mice. The novel finding that low-impact loading could counter detrimental disuse effects in the perlecan-deficient skeleton suggests a strategy to maintain skeletal health in SJS patients.
Collapse
Affiliation(s)
- Ashutosh Parajuli
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA; (A.P.); (S.P.); (J.R.M.); (X.L.L.)
| | - Shaopeng Pei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA; (A.P.); (S.P.); (J.R.M.); (X.L.L.)
| | - Hongbo Zhao
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.Z.)
| | - Jerahme R. Martinez
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA; (A.P.); (S.P.); (J.R.M.); (X.L.L.)
| | - X. Lucas Lu
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA; (A.P.); (S.P.); (J.R.M.); (X.L.L.)
| | - X. Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.Z.)
| | - Mary C. Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center, Houston, TX 77054, USA;
| | - Catherine B. Kirn-Safran
- Department of Biology, Widener University, Chester, PA 19013, USA;
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Liyun Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA; (A.P.); (S.P.); (J.R.M.); (X.L.L.)
- Correspondence: ; Tel.: +1-302-831-2659
| |
Collapse
|
49
|
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
|
50
|
Hagan ML, Yu K, Zhu J, Vinson BN, Roberts RL, Montesinos Cartagena M, Johnson MH, Wang L, Isales CM, Hamrick MW, McNeil PL, McGee‐Lawrence ME. Decreased pericellular matrix production and selection for enhanced cell membrane repair may impair osteocyte responses to mechanical loading in the aging skeleton. Aging Cell 2020; 19:e13056. [PMID: 31743583 PMCID: PMC6974724 DOI: 10.1111/acel.13056] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/16/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Transient plasma membrane disruptions (PMD) occur in osteocytes with in vitro and in vivo loading, initiating mechanotransduction. The goal here was to determine whether osteocyte PMD formation or repair is affected by aging. Osteocytes from old (24 months) mice developed fewer PMD (-76% females, -54% males) from fluid shear than young (3 months) mice, and old mice developed fewer osteocyte PMD (-51%) during treadmill running. This was due at least in part to decreased pericellular matrix production, as studies revealed that pericellular matrix is integral to formation of osteocyte PMD, and aged osteocytes produced less pericellular matrix (-55%). Surprisingly, osteocyte PMD repair rate was faster (+25% females, +26% males) in osteocytes from old mice, and calcium wave propagation to adjacent nonwounded osteocytes was blunted, consistent with impaired mechanotransduction downstream of PMD in osteocytes with fast PMD repair in previous studies. Inducing PMD via fluid flow in young osteocytes in the presence of oxidative stress decreased postwounding cell survival and promoted accelerated PMD repair in surviving cells, suggesting selective loss of slower-repairing osteocytes. Therefore, as oxidative stress increases during aging, slower-repairing osteocytes may be unable to successfully repair PMD, leading to slower-repairing osteocyte death in favor of faster-repairing osteocyte survival. Since PMD are an important initiator of mechanotransduction, age-related decreases in pericellular matrix and loss of slower-repairing osteocytes may impair the ability of bone to properly respond to mechanical loading with bone formation. These data suggest that PMD formation and repair mechanisms represent new targets for improving bone mechanosensitivity with aging.
Collapse
Affiliation(s)
- Mackenzie L. Hagan
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
| | - Kanglun Yu
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
| | - Jiali Zhu
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
| | - Brooke N. Vinson
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
| | - Rachel L. Roberts
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
| | | | - Maribeth H. Johnson
- Department of Neuroscience and Regenerative MedicineAugusta UniversityAugustaGA
| | - Liyun Wang
- Department of Mechanical EngineeringUniversity of DelawareNewarkDE
| | - Carlos M. Isales
- Department of Neuroscience and Regenerative MedicineAugusta UniversityAugustaGA
| | - Mark W. Hamrick
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
| | - Paul L. McNeil
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
| | - Meghan E. McGee‐Lawrence
- Department of Cellular Biology and AnatomyMedical College of GeorgiaAugusta UniversityAugustaGA
- Department of Orthopaedic SurgeryAugusta UniversityAugustaGA
| |
Collapse
|