201
|
Tomlinson RE, Schmieder AH, Quirk JD, Lanza GM, Silva MJ. Antagonizing the αv β3 integrin inhibits angiogenesis and impairs woven but not lamellar bone formation induced by mechanical loading. J Bone Miner Res 2014; 29:1970-80. [PMID: 24644077 PMCID: PMC4323187 DOI: 10.1002/jbmr.2223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 02/25/2014] [Accepted: 02/27/2014] [Indexed: 12/26/2022]
Abstract
Angiogenesis and osteogenesis are critically linked, although the role of angiogenesis is not well understood in osteogenic mechanical loading. In this study, either damaging or non-damaging cyclic axial compression was used to generate woven bone formation (WBF) or lamellar bone formation (LBF), respectively, at the mid-diaphysis of the adult rat forelimb. αv β3 integrin-targeted nanoparticles or vehicle was injected intravenously after mechanical loading. β3 integrin subunit expression on vasculature was maximal 7 days after damaging mechanical loading, but was still robustly expressed 14 days after loading. Accordingly, targeted nanoparticle delivery in WBF-loaded limbs was increased compared with non-loaded limbs. Vascularity was dramatically increased after WBF loading (+700% on day 14) and modestly increased after LBF loading (+50% on day 14). This increase in vascularity was inhibited by nanoparticle treatment in both WBF- and LBF-loaded limbs at days 7 and 14 after loading. Decreased vascularity led to diminished woven, but not lamellar, bone formation. Decreased woven bone formation resulted in impaired structural properties of the skeletal repair, particularly in post-yield behavior. These results demonstrate that αv β3 integrin-mediated angiogenesis is critical for recovering fracture resistance after bone injury but is not required for bone modeling after modest mechanical strain. © 2014 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ryan E. Tomlinson
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University in St. Louis, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Anne H. Schmieder
- Department of Medicine, Division of Cardiology, Washington University in St. Louis, Saint Louis, MO, USA
| | - James D. Quirk
- Department of Radiology, Washington University in St. Louis, Saint Louis, MO, USA
| | - Gregory M. Lanza
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
- Department of Medicine, Division of Cardiology, Washington University in St. Louis, Saint Louis, MO, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University in St. Louis, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| |
Collapse
|
202
|
Mechanotransduction in musculoskeletal tissue regeneration: effects of fluid flow, loading, and cellular-molecular pathways. BIOMED RESEARCH INTERNATIONAL 2014; 2014:863421. [PMID: 25215295 PMCID: PMC4151828 DOI: 10.1155/2014/863421] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/13/2014] [Indexed: 12/28/2022]
Abstract
While mechanotransductive signal is proven essential for tissue regeneration, it is critical to determine specific cellular responses to such mechanical signals and the underlying mechanism. Dynamic fluid flow induced by mechanical loading has been shown to have the potential to regulate bone adaptation and mitigate bone loss. Mechanotransduction pathways are of great interests in elucidating how mechanical signals produce such observed effects, including reduced bone loss, increased bone formation, and osteogenic cell differentiation. The objective of this review is to develop a molecular understanding of the mechanotransduction processes in tissue regeneration, which may provide new insights into bone physiology. We discussed the potential for mechanical loading to induce dynamic bone fluid flow, regulation of bone adaptation, and optimization of stimulation parameters in various loading regimens. The potential for mechanical loading to regulate microcirculation is also discussed. Particularly, attention is allotted to the potential cellular and molecular pathways in response to loading, including osteocytes associated with Wnt signaling, elevation of marrow stem cells, and suppression of adipotic cells, as well as the roles of LRP5 and microRNA. These data and discussions highlight the complex yet highly coordinated process of mechanotransduction in bone tissue regeneration.
Collapse
|
203
|
Predicting and validating the pathway of Wnt3a-driven suppression of osteoclastogenesis. Cell Signal 2014; 26:2358-69. [PMID: 25038457 DOI: 10.1016/j.cellsig.2014.07.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 06/03/2014] [Accepted: 07/10/2014] [Indexed: 11/23/2022]
Abstract
Wnt signaling plays a major role in bone homeostasis and mechanotransduction, but its role and regulatory mechanism in osteoclast development are not fully understood. Through genome-wide in silico analysis, we examined Wnt3a-driven regulation of osteoclast development. Mouse bone marrow-derived cells were incubated with RANKL in the presence and absence of Wnt3a. Using microarray mRNA expression data, we conducted principal component analysis and predicted transcription factor binding sites (TFBSs) that were potentially involved in the responses to RANKL and Wnt3a. The principal component analysis predicted potential Wnt3a responsive regulators that would reverse osteoclast development, and a TFBS prediction algorithm indicated that the AP1 binding site would be linked to Wnt3a-driven suppression. Since c-Fos was upregulated by RANKL and downregulated by Wnt3a in a dose-dependent manner, we examined its role using RNA interference. The partial silencing of c-Fos suppressed RANKL-driven osteoclastogenesis by downregulating NFATc1, a master transcription factor of osteoclast development. Although the involvement of c-Myc was predicted and partially silencing c-Myc slightly reduced the level of TRAP, c-Myc silencing did not alter the expression of NFATc1. Collectively, the presented systems-biology approach demonstrates that Wnt3a attenuates RANKL-driven osteoclastogenesis by blocking c-Fos expression and suggests that mechanotransduction of bone alters the development of not only osteoblasts but also osteoclasts through Wnt signaling.
Collapse
|
204
|
Ozcivici E, Zhang W, Donahue LR, Judex S. Quantitative trait loci that modulate trabecular bone's risk of failure during unloading and reloading. Bone 2014; 64:25-32. [PMID: 24698783 DOI: 10.1016/j.bone.2014.03.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 02/01/2014] [Accepted: 03/22/2014] [Indexed: 01/23/2023]
Abstract
Genetic makeup of an individual is a strong determinant of the morphologic and mechanical properties of bone. Here, in an effort to identify quantitative trait loci (QTLs) for changes in the simulated mechanical parameters of trabecular bone during altered mechanical demand, we subjected 352 second generation female adult (16 weeks old) BALBxC3H mice to 3 weeks of hindlimb unloading followed by 3 weeks of reambulation. Longitudinal in vivo microcomputed tomography (μCT) scans tracked trabecular changes in the distal femur. Tomographies were directly translated into finite element (FE) models and subjected to a uniaxial compression test. Apparent trabecular stiffness and components of the Von Mises (VM) stress distributions were computed for the distal metaphysis and associated with QTLs. At baseline, five QTLs explained 20% of the variation in trabecular peak stresses across the mouse population. During unloading, three QTLs accounted for 14% of the variability in peak stresses. During reambulation, one QTL accounted for 5% of the variability in peak stresses. QTLs were also identified for mechanically induced changes in stiffness, median stress values and skewness of stress distributions. There was little overlap between QTLs identified for baseline and QTLs for longitudinal changes in mechanical properties, suggesting that distinct genes may be responsible for the mechanical response of trabecular bone. Unloading related QTLs were also different from reambulation related QTLs. Further, QTLs identified here for mechanical properties differed from previously identified QTLs for trabecular morphology, perhaps revealing novel gene targets for reducing fracture risk in individuals exposed to unloading and for maximizing the recovery of trabecular bone's mechanical properties during reambulation.
Collapse
Affiliation(s)
- Engin Ozcivici
- Department of Mechanical Engineering, Izmir Institute of Technology, Urla, Izmir 35430, Turkey.
| | | | | | - Stefan Judex
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
205
|
|
206
|
Wang L, Foster BL, Kram V, Nociti FH, Zerfas PM, Tran AB, Young MF, Somerman MJ. Fibromodulin and Biglycan Modulate Periodontium through TGFβ/BMP Signaling. J Dent Res 2014; 93:780-7. [PMID: 24966230 DOI: 10.1177/0022034514541126] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/02/2014] [Indexed: 12/15/2022] Open
Abstract
A full understanding of the key regulators controlling periodontal development and homeostasis is necessary for the design of improved periodontal regenerative therapies. Small leucine-rich proteoglycans (SLRPs) are extracellular matrix molecules suggested to regulate collagen organization and cell signaling. Mice with double-deficiency of 2 SLRPs, fibromodulin and biglycan (dKO), acquire skeletal abnormalities, but their roles in regulating the periodontium remain undefined and were the focus of our studies. Transmission electron microscopy studies showed abnormal collagen fibrils in the periodontal ligament (PDL) and altered remodeling of alveolar bone in dKO mice. Immunohistochemistry (IHC) revealed increased staining of SLRPs (asporin, lumican, and decorin) and dentin matrix protein-1 (DMP1, a mechanosensory/osteocyte marker), while osteoblast markers, bone sialoprotein and osteopontin, remained unchanged. Disruption of homeostasis was further evidenced by increased expression of receptor-activator of nuclear factor-κB ligand (RANKL) and elevated numbers of osteoclasts, especially noted around the alveolar bone of molars (buccal side) and incisors. Polymerase chain reaction (PCR) array revealed hyperactive transforming growth factors beta/bone morphogenetic protein (TGFβ/BMP) signaling in dKO PDL tissues, which was further confirmed by elevated expression of phosphorylated Smad5 (p-Smad5) by IHC in dKO PDL. These studies highlight the importance of SLRPs in maintaining periodontal homeostasis through regulation of TGFβ/BMP signaling, matrix turnover, and collagen organization.
Collapse
Affiliation(s)
- L Wang
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - B L Foster
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - V Kram
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - F H Nociti
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA State University of Campinas, Piracicaba Dental School, Department of Prosthodontics and Periodontics, Piracicaba, Brazil
| | - P M Zerfas
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health (NIH), Bethesda, MD, USA
| | - A B Tran
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M F Young
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
207
|
Abstract
Bone defects requiring grafts to promote healing are frequently occurring and costly problems in health care. Chitosan, a biodegradable, naturally occurring polymer, has drawn considerable attention in recent years as scaffolding material in tissue engineering and regenerative medicine. Chitosan is especially attractive as a bone scaffold material because it supports the attachment and proliferation of osteoblast cells as well as formation of mineralized bone matrix. In this review, we discuss the fundamentals of bone tissue engineering and the unique properties of chitosan as a scaffolding material to treat bone defects for hard tissue regeneration. We present the common methods for fabrication and characterization of chitosan scaffolds, and discuss the influence of material preparation and addition of polymeric or ceramic components or biomolecules on chitosan scaffold properties such as mechanical strength, structural integrity, and functional bone regeneration. Finally, we highlight recent advances in development of chitosan-based scaffolds with enhanced bone regeneration capability.
Collapse
Affiliation(s)
- Sheeny Lan Levengood
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195 USA
| | - Miqin Zhang
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195 USA
| |
Collapse
|
208
|
Mechanical loading in osteocytes induces formation of a Src/Pyk2/MBD2 complex that suppresses anabolic gene expression. PLoS One 2014; 9:e97942. [PMID: 24841674 PMCID: PMC4026426 DOI: 10.1371/journal.pone.0097942] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/25/2014] [Indexed: 11/19/2022] Open
Abstract
Mechanical stimulation of the skeleton promotes bone gain and suppresses bone loss, ultimately resulting in improved bone strength and fracture resistance. The molecular mechanisms directing anabolic and/or anti-catabolic actions on the skeleton during loading are not fully understood. Identifying molecular mechanisms of mechanotransduction (MTD) signaling cascades could identify new therapeutic targets. Most research into MTD mechanisms is typically focused on understanding the signaling pathways that stimulate new bone formation in response to load. However, we investigated the structural, signaling and transcriptional molecules that suppress the stimulatory effects of loading. The high bone mass phenotype of mice with global deletion of either Pyk2 or Src suggests a role for these tyrosine kinases in repression of bone formation. We used fluid shear stress as a MTD stimulus to identify a novel Pyk2/Src-mediated MTD pathway that represses mechanically-induced bone formation. Our results suggest Pyk2 and Src function as molecular switches that inhibit MTD in our mechanically stimulated osteocyte culture experiments. Once activated by oscillatory fluid shear stress (OFSS), Pyk2 and Src translocate to and accumulate in the nucleus, where they associate with a protein involved in DNA methylation and the interpretation of DNA methylation patterns –methyl-CpG-binding domain protein 2 (MBD2). OFSS-induced Cox-2 and osteopontin expression was enhanced in Pyk2 KO osteoblasts, while inhibition of Src enhanced osteocalcin expression in response to OFSS. We found that Src kinase activity increased in the nucleus of osteocytes in response to OFSS and an interaction activated between Src (Y418) and Pyk2 (Y402) increased in response to OFSS. Thus, as a mechanism to prevent an over-reaction to physical stimulation, mechanical loading may induce the formation of a Src/Pyk2/MBD2 complex in the nucleus that functions to suppress anabolic gene expression.
Collapse
|
209
|
Xu L, Wang X, Zhang Q, Yang W, Zhu W, Zhao K. Immediate versus early loading of flapless placed dental implants: a systematic review. J Prosthet Dent 2014; 112:760-9. [PMID: 24831750 DOI: 10.1016/j.prosdent.2014.01.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 01/27/2014] [Accepted: 01/27/2014] [Indexed: 10/25/2022]
Abstract
STATEMENT OF PROBLEM The flapless implant technique is a predictable procedure with several advantages and a high overall implant survival rate. Immediate loading and early loading have been widely used in dental implant therapies and provide improved esthetics, with enhanced function and comfort. However, the scientific support for immediate or early loading approaches for flapless-placed dental implants is unclear. PURPOSE The purpose of this systematic review was to assess the effectiveness and safety of the immediate versus early loading of dental implants with flapless placement. MATERIAL AND METHODS The Cochrane Central Register of Controlled Trials, MEDLINE, EMBASE, CNKI database, VIP database, WANFANG Database, and World Health Organization International Clinical Trials Registry Platform Search Portal were searched (up to October 2012). The systematic review included clinical randomized controlled trials that compared immediate with early loading of flapless-placed dental implants to replace missing teeth in adult participants who were partially or completely edentulous. The selection of included studies, data extraction, and assessment of the quality of the studies and evidence were conducted independently by 2 reviewers. RESULTS Six articles that reported on 4 randomized controlled trials that involved 180 selected participants were included. The implant failure rate was from 0.0% to 3.3% in both immediate and early loading groups with flapless implantation. No statistically significant differences were found in implant failure rates, periimplant marginal bone-level changes, or complications between the 2 groups. More participants preferred immediate loading rather than waiting for nearly 2 months. CONCLUSIONS Within the limitation of needing additional high-quality evidence, immediate and early loading of dental implants after flapless placement both demonstrated an acceptable short- to medium-term survival rate. Immediate loading seems more acceptable because of the time benefit.
Collapse
Affiliation(s)
- Lin Xu
- Resident, Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaodong Wang
- Resident, Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qin Zhang
- Resident, Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wen Yang
- Resident, Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenjun Zhu
- Resident, Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ke Zhao
- Professor, Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
210
|
Chen AB, Zhang P, Yokota H. Evaluating treatment of osteoporosis using particle swarm on a bone remodelling mathematical model. IET Syst Biol 2014; 7:231-42. [PMID: 24712100 DOI: 10.1049/iet-syb.2013.0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bone loss in osteoporosis, commonly observed in postmenopausal women and the elderly, is caused by an imbalance in activities of bone-forming osteoblasts and bone-resorbing osteoclasts. To treat osteoporosis and increase bone mineral density (BMD), physical activities and drugs are often recommended. Complex systems dynamics prevent an intuitive prediction of treatment strategies, and little is known about an optimal sequence for the combinatorial use of available treatments. In this study, the authors built a mathematical model of bone remodelling and developed a treatment strategy for mechanical loading and salubrinal, a synthetic chemical agent that enhances bone formation and prevents bone resorption. The model formulated a temporal BMD change of a mouse's whole skeleton in response to ovariectomy, mechanical loading and administration of salubrinal. Particle swarm optimisation was employed to maximise a performance index (a function of BMD and treatment cost) to find an ideal sequence of treatment. The best treatment was found to start with mechanical loading followed by salubrinal. As treatment costs increased, the sequence started with no treatment and usage of salubrinal became scarce. The treatment strategy will depend on individual needs and costs, and the proposed model is expected to contribute to the development of personalised treatment strategies.
Collapse
|
211
|
Knapik DM, Perera P, Nam J, Blazek AD, Rath B, Leblebicioglu B, Das H, Wu LC, Hewett TE, Agarwal SK, Robling AG, Flanigan DC, Lee BS, Agarwal S. Mechanosignaling in bone health, trauma and inflammation. Antioxid Redox Signal 2014; 20:970-85. [PMID: 23815527 PMCID: PMC3924811 DOI: 10.1089/ars.2013.5467] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
SIGNIFICANCE Mechanosignaling is vital for maintaining the structural integrity of bone under physiologic conditions. These signals activate and suppress multiple signaling cascades regulating bone formation and resorption. Understanding these pathways is of prime importance to exploit their therapeutic potential in disorders associated with bone loss due to disuse, trauma, or disruption of homeostatic mechanisms. RECENT ADVANCES In the case of cells of the bone, an impressive amount of data has been generated that provides evidence of a complex mechanism by which mechanical signals can maintain or disrupt cellular homeostasis by driving transcriptional regulation of growth factors, matrix proteins and inflammatory mediators in health and inflammation. Mechanical signals act on cells in a magnitude dependent manner to induce bone deposition or resorption. During health, physiological levels of these signals are essential for maintaining bone strength and architecture, whereas during inflammation, similar signals can curb inflammation by suppressing the nuclear factor kappa B (NF-κB) signaling cascade, while upregulating matrix synthesis via mothers against decapentaplegic homolog and/or Wnt signaling cascades. Contrarily, excessive mechanical forces can induce inflammation via activation of the NF-κB signaling cascade. CRITICAL ISSUES Given the osteogenic potential of mechanical signals, it is imperative to exploit their therapeutic efficacy for the treatment of bone disorders. Here we review select signaling pathways and mediators stimulated by mechanical signals to modulate the strength and integrity of the bone. FUTURE DIRECTIONS Understanding the mechanisms of mechanotransduction and its effects on bone lay the groundwork for development of nonpharmacologic mechanostimulatory approaches for osteodegenerative diseases and optimal bone health.
Collapse
Affiliation(s)
- Derrick M Knapik
- 1 Department of Orthopaedic Surgery, The Ohio State University College of Medicine , Columbus, Ohio
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Colaianni G, Cuscito C, Mongelli T, Oranger A, Mori G, Brunetti G, Colucci S, Cinti S, Grano M. Irisin enhances osteoblast differentiation in vitro. Int J Endocrinol 2014; 2014:902186. [PMID: 24723951 PMCID: PMC3960733 DOI: 10.1155/2014/902186] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/13/2014] [Indexed: 12/15/2022] Open
Abstract
It has been recently demonstrated that exercise activity increases the expression of the myokine Irisin in skeletal muscle, which is able to drive the transition of white to brown adipocytes, likely following a phenomenon of transdifferentiation. This new evidence supports the idea that muscle can be considered an endocrine organ, given its ability to target adipose tissue by promoting energy expenditure. In accordance with these new findings, we hypothesized that Irisin is directly involved in bone metabolism, demonstrating its ability to increase the differentiation of bone marrow stromal cells into mature osteoblasts. Firstly, we confirmed that myoblasts from mice subjected to 3 weeks of free wheel running increased Irisin expression compared to nonexercised state. The conditioned media (CM) collected from myoblasts of exercised mice induced osteoblast differentiation in vitro to a greater extent than those of mice housed in resting conditions. Furthermore, the differentiated osteoblasts increased alkaline phosphatase and collagen I expression by an Irisin-dependent mechanism. Our results show, for the first time, that Irisin directly targets osteoblasts, enhancing their differentiation. This finding advances notable perspectives in future studies which could satisfy the ongoing research of exercise-mimetic therapies with anabolic action on the skeleton.
Collapse
Affiliation(s)
- Graziana Colaianni
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
| | - Concetta Cuscito
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
| | - Teresa Mongelli
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
| | - Angela Oranger
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
| | - Silvia Colucci
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, United Hospitals—University of Ancona, 60020 Ancona, Italy
| | - Maria Grano
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124 Bari, Italy
- *Maria Grano:
| |
Collapse
|
213
|
Kang KS, Robling AG. New Insights into Wnt-Lrp5/6-β-Catenin Signaling in Mechanotransduction. Front Endocrinol (Lausanne) 2014; 5:246. [PMID: 25653639 PMCID: PMC4299511 DOI: 10.3389/fendo.2014.00246] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/31/2014] [Indexed: 11/13/2022] Open
Abstract
Mechanical loading is essential to maintain normal bone metabolism and the balance between bone formation and resorption. The cellular mechanisms that control mechanotransduction are not fully defined, but several key pathways have been identified. We discuss the roles of several components of the Wnt signaling cascade, namely Lrp5, Lrp6, and β-catenin in mechanical loading-induced bone formation. Lrp5 is an important Wnt co-receptor for regulating bone mass and mechanotransduction, and appears to function principally by augmenting bone formation. Lrp6 also regulates bone mass but its action might involve resorption as well as formation. The role of Lrp6 in mechanotransduction is unclear. Studies addressing the role of β-catenin in bone metabolism and mechanotransduction highlight the uncertainties in downstream modulators of Lrp5 and Lrp6. Taken together, these data indicate that mechanical loading might affect bone regulation triggering the canonical Wnt signaling (and perhaps other pathways) not only via Lrp5 but also via Lrp6. Further work is needed to clarify the role of the Wnt signaling pathway in Lrp5 and/or Lrp6-mediated mechanotransduction, which could eventually lead to powerful therapeutic agents that might mimic the anabolic effects of mechanical stimulation.
Collapse
Affiliation(s)
- Kyung Shin Kang
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander G. Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Engineering, Indiana University–Purdue University at Indianapolis, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
- *Correspondence: Alexander G. Robling, Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 5035, Indianapolis, IN 46202, USA e-mail:
| |
Collapse
|
214
|
Jing D, Baik AD, Lu XL, Zhou B, Lai X, Wang L, Luo E, Guo XE. In situ intracellular calcium oscillations in osteocytes in intact mouse long bones under dynamic mechanical loading. FASEB J 2013; 28:1582-92. [PMID: 24347610 DOI: 10.1096/fj.13-237578] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Osteocytes have been hypothesized to be the major mechanosensors in bone. How in situ osteocytes respond to mechanical stimuli is still unclear because of technical difficulties. In vitro studies have shown that osteocytes exhibited unique calcium (Ca(2+)) oscillations to fluid shear. However, whether this mechanotransduction phenomenon holds for in situ osteocytes embedded within a mineralized bone matrix under dynamic loading remains unknown. Using a novel synchronized loading/imaging technique, we successfully visualized in real time and quantified Ca(2+) responses in osteocytes and bone surface cells in situ under controlled dynamic loading on intact mouse tibia. The resultant fluid-induced shear stress on the osteocyte in the lacunocanalicular system (LCS) was also quantified. Osteocytes, but not surface cells, displayed repetitive Ca(2+) spikes in response to dynamic loading, with spike frequency and magnitude dependent on load magnitude, tissue strain, and shear stress in the LCS. The Ca(2+) oscillations were significantly reduced by endoplasmic reticulum (ER) depletion and P2 purinergic receptor (P2R)/phospholipase C (PLC) inhibition. This study provides direct evidence that osteocytes respond to in situ mechanical loading by Ca(2+) oscillations, which are dependent on the P2R/PLC/inositol trisphosphate/ER pathway. This study develops a novel approach in skeletal mechanobiology and also advances our fundamental knowledge of bone mechanotransduction.
Collapse
Affiliation(s)
- Da Jing
- 1351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Ave., Columbia University, New York, NY 10027, USA.
| | | | | | | | | | | | | | | |
Collapse
|
215
|
SHANG PENG, ZHANG JIAN, QIAN AIRONG, LI JINGBAO, MENG RUI, DI SHENGMENG, HU LIFANG, GU ZHONGZE. BONE CELLS UNDER MICROGRAVITY. J MECH MED BIOL 2013. [DOI: 10.1142/s021951941340006x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Weightlessness environment (also microgravity) during the exploration of space is the major condition which must be faced by astronauts. One of the most serious adverse effects on astronauts is the weightlessness-induced bone loss due to the unbalanced bone remodeling. Bone remodeling of human beings has evolved during billions of years to make bone tissue adapt to the gravitational field of Earth (1g) and maintain skeleton structure to meet mechanical loading on Earth. However, under weightlessness environment the skeleton system no longer functions against the pull of gravity, so there is no necessity to keep bone strong enough to support the body's weight. Therefore, the balance of bone remodeling is disrupted and bone loss occurs, which is extremely deleterious to an astronaut's health during long-term spaceflight. Bone remodeling is mainly orchestrated by bone mesenchymal stem cells, osteoblasts, osteocytes, and osteoclasts. Here, we review how these bone cells respond to microgravity environment.
Collapse
Affiliation(s)
- PENG SHANG
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, P. R. China
- The State Key Laboratory of Bioelectonics, Southeast University, 210096, P. R. China
| | - JIAN ZHANG
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, P. R. China
| | - AIRONG QIAN
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, P. R. China
| | - JINGBAO LI
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, P. R. China
| | - RUI MENG
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, P. R. China
| | - SHENGMENG DI
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, P. R. China
| | - LIFANG HU
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, P. R. China
| | - ZHONGZE GU
- The State Key Laboratory of Bioelectonics, Southeast University, 210096, P. R. China
| |
Collapse
|
216
|
Shu L, Zhang H, Boyce B, Xing L. Ubiquitin E3 ligase Wwp1 negatively regulates osteoblast function by inhibiting osteoblast differentiation and migration. J Bone Miner Res 2013; 28:1925-35. [PMID: 23553732 PMCID: PMC3749248 DOI: 10.1002/jbmr.1938] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 03/01/2013] [Accepted: 03/20/2013] [Indexed: 01/09/2023]
Abstract
Ubiquitin E3 ligase-mediated protein degradation promotes proteasomal degradation of key positive regulators of osteoblast functions. For example, the E3 ligases--SMAD-specific E3 ubiquitin protein ligase 1 (Smurf1), Itch, and WW domain-containing E3 ubiquitin protein ligase 1 (Wwp1)--promote degradation of Runt-related transcription factor 2 (Runx2), transcription factor jun-B (JunB), and chemokine (C-X-C) receptor type 4 (CXCR-4) proteins to inhibit their functions. However, the role of E3 ligases in age-associated bone loss is unknown. We found that the expression level of Wwp1, but not Smurf1 or Itch, was significantly increased in CD45-negative (CD45(-)) bone marrow-derived mesenchymal stem cells from 6-month-old and 12-month-old wild-type (WT) mice. Wwp1 knockout (Wwp1(-/-)) mice developed increased bone mass as they aged, associated with increased bone formation rates and normal bone resorption parameters. Bone marrow stromal cells (BMSCs) from Wwp1(-/-) mice formed increased numbers and areas of alkaline phosphatase(+) and Alizarin red(+) nodules and had increased migration potential toward chemokine (C-X-C motif) ligand 12 (CXCL12) gradients. Runx2, JunB, and CXCR-4 protein levels were significantly increased in Wwp1(-/-) BMSCs. Wwp1(-/-) BMSCs had increased amount of ubiquitinated JunB protein, but Runx2 ubiquitination was no change. Knocking down JunB in Wwp1(-/-) BMSCs returned Runx2 protein levels to that in WT cells. Thus, Wwp1 negatively regulates osteoblast functions by affecting both their migration and differentiation. Mechanisms designed to decrease Wwp1 levels in BMSCs may represent a new approach to prevent the decrease in osteoblastic bone formation associated with aging.
Collapse
Affiliation(s)
| | | | | | - Lianping Xing
- Correspondence to: Lianping Xing, Department of Pathology and Laboratory Medicine, 601 Elmwood Ave, Box 626, Rochester, NY 14642, USA. Phone (585) 273-4090, Fax (585) 756-4468,
| |
Collapse
|
217
|
Wang H, Brennan TA, Russell E, Kim JH, Egan KP, Chen Q, Israelite C, Schultz DC, Johnson FB, Pignolo RJ. R-Spondin 1 promotes vibration-induced bone formation in mouse models of osteoporosis. J Mol Med (Berl) 2013; 91:1421-9. [PMID: 23974989 DOI: 10.1007/s00109-013-1068-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 06/12/2013] [Accepted: 06/17/2013] [Indexed: 12/29/2022]
Abstract
UNLABELLED Bone tissue adapts to its functional environment by optimizing its morphology for mechanical demand. Among the mechanosensitive cells that recognize and respond to forces in the skeleton are osteocytes, osteoblasts, and mesenchymal progenitor cells (MPCs). Therefore, the ability to use mechanical signals to improve bone health through exercise and devices that deliver mechanical signals is an attractive approach to age-related bone loss; however, the extracellular or circulating mediators of such signals are largely unknown. Using SDS-PAGE separation of proteins secreted by MPCs in response to low-magnitude mechanical signals and in-gel trypsin digestion followed by HPLC and mass spectroscopy, we identified secreted proteins up-regulated by vibratory stimulation. We exploited a cell senescence-associated secretory phenotype screen and reasoned that a subset of vibration-induced proteins with diminished secretion by senescent MPCs will have the capacity to promote bone formation in vivo. We identified one such vibration-induced bone-enhancing (vibe) gene as R-spondin 1, a Wnt pathway modulator, and demonstrated that it has the capacity to promote bone formation in three mouse models of age-related bone loss. By virtue of their secretory status, some vibe proteins may be candidates for pre-clinical development as anabolic agents for the treatment of osteoporosis. KEY MESSAGE Mesenchymal stem cells respond to low magnitude mechanical signals (vibration). R-Spondin 1 is upregulated by mechanical signals and secreted. R-Spondin 1 promotes bone formation in three mouse models of osteoporosis.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Sachs N, Sonnenberg A. Cell-matrix adhesion of podocytes in physiology and disease. Nat Rev Nephrol 2013; 9:200-10. [PMID: 23338211 DOI: 10.1038/nrneph.2012.291] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cell-matrix adhesion is crucial for maintaining the mechanical integrity of epithelial tissues. Podocytes--a key component of the glomerular filtration barrier--are exposed to permanent transcapillary filtration pressure and must therefore adhere tightly to the underlying glomerular basement membrane (GBM). The major cell-matrix adhesion receptor in podocytes is the integrin α3β1, which connects laminin 521 in the GBM through various adaptor proteins to the intracellular actin cytoskeleton. Other cell-matrix adhesion receptors expressed by podocytes include the integrins α2β1 and αvβ3, α-dystroglycan, syndecan-4 and type XVII collagen. Mutations in genes encoding any of the components critical for podocyte adhesion cause glomerular disease. This Review highlights recent advances in our understanding of the cell biology and genetics of podocyte adhesion with special emphasis on glomerular disease.
Collapse
Affiliation(s)
- Norman Sachs
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
219
|
Abstract
Skeletal health is dependent on the balance between bone resorption and formation during bone remodeling. Multiple signaling pathways play essential roles in the maintenance of skeletal integrity by positively or negatively regulating bone cells. During the last years, significant advances have been made in our understanding of the essential signaling pathways that regulate bone cell commitment, differentiation and survival. New signaling anabolic pathways triggered by parathyroid hormone, local growth factors, Wnt signaling, and calcium sensing receptor have been identified. Novel signals induced by interactions between bone cells-matrix (integrins), osteoblasts/osteocytes (cadherins, connexins), and osteoblasts/osteoclast (ephrins, Wnt-RhoA, semaphorins) have been discovered. Recent studies revealed the key pathways (MAPK, PI3K/Akt) that critically control bone cells and skeletal mass. This review summarizes the most recent knowledge on the major signaling pathways that control bone cells, and their potential impact on the development of therapeutic strategies to improve human bone health.
Collapse
Affiliation(s)
- Pierre J Marie
- Laboratory of osteoblast biology and pathology, INSERM, UMR-606, University Paris Diderot, Sorbonne Paris Cité, Hopital Lariboisiere, 2 rue Ambroise Pare, 75475 Paris cedex 10, France.
| |
Collapse
|
220
|
Grethen E, Hill KM, Jones R, Cacucci BM, Gupta CE, Acton A, Considine RV, Peacock M. Serum leptin, parathyroid hormone, 1,25-dihydroxyvitamin D, fibroblast growth factor 23, bone alkaline phosphatase, and sclerostin relationships in obesity. J Clin Endocrinol Metab 2012; 97:1655-62. [PMID: 22362819 PMCID: PMC3339883 DOI: 10.1210/jc.2011-2280] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Obesity is associated with hyperparathyroidism and increased bone mass and turnover, but their pathogeneses are unclear. AIMS Our aim was to determine in obesity interrelationships among serum levels of leptin, the mineral-regulating hormones, bone turnover markers, and sclerostin. METHODS This case-control study was performed in 20 women having bariatric surgery and 20 control women matched for race and age. Anthropometrics and fasting serum biochemistries were measured in controls and in bariatric patients the morning of surgery. RESULTS Body mass index (48.9 vs. 25.4 kg/m(2)), weight (128.6 vs. 71.9 kg), serum leptin (74.6 vs. 25.2 ng/ml), PTH (44.5 vs. 28.8 pg/ml), fibroblast growth factor 23 (FGF23) (42.4 vs. 25.9 pg/ml), and bone alkaline phosphatase (BAP) (25.8 vs. 17.5 U/liter) were higher, but height (162.3 vs. 167.7 cm) and 1,25-dihydroxyvitamin D (1,25D) (39.2 vs. 48.7 pg/ml) were lower in bariatric surgery patients than controls. There was no difference in serum sclerostin, amino-terminal collagen cross-links, 25-hydroxyvitamin D (25D), calcium, phosphate, and creatinine between groups. In the combined sample, leptin was positively related to PTH, FGF23, and BAP but not to 1,25D or sclerostin. Multiple regression analysis demonstrated that PTH was predicted by leptin and Ca (R(2) = 0.39); 1,25D by 25D, FGF23, and phosphate (R(2) = 0.43); FGF23 by leptin and 1,25D (R(2) = 0.27); BAP by leptin, PTH, and Ca (R(2) = 0.39); and sclerostin by leptin and PTH (R(2) = 0.20). CONCLUSIONS Women having bariatric surgery had higher leptin, PTH, FGF23, and BAP and lower 1,25D than controls. Leptin predicted the serum levels of PTH, 1,25D, and FGF23, the mineral-regulating hormones, and BAP, a bone formation marker, in women with body mass index ranging from 13.9-65.8 kg/m(2). The results suggest that leptin has an endocrine or paracrine effect on PTH and FGF23 production and that PTH may be one of the signals in obesity that leads to increased bone mass.
Collapse
Affiliation(s)
- Elizabeth Grethen
- Division of Endocrinology and Metabolism, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
221
|
Leong NL, Hurng JM, Djomehri SI, Gansky SA, Ryder MI, Ho SP. Age-related adaptation of bone-PDL-tooth complex: Rattus-Norvegicus as a model system. PLoS One 2012; 7:e35980. [PMID: 22558292 PMCID: PMC3340399 DOI: 10.1371/journal.pone.0035980] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/26/2012] [Indexed: 01/18/2023] Open
Abstract
Functional loads on an organ induce tissue adaptations by converting mechanical energy into chemical energy at a cell-level. The transducing capacity of cells alters physico-chemical properties of tissues, developing a positive feedback commonly recognized as the form-function relationship. In this study, organ and tissue adaptations were mapped in the bone-tooth complex by identifying and correlating biomolecular expressions to physico-chemical properties in rats from 1.5 to 15 months. However, future research using hard and soft chow over relevant age groups would decouple the function related effects from aging affects. Progressive curvature in the distal root with increased root resorption was observed using micro X-ray computed tomography. Resorption was correlated to the increased activity of multinucleated osteoclasts on the distal side of the molars until 6 months using tartrate resistant acid phosphatase (TRAP). Interestingly, mononucleated TRAP positive cells within PDL vasculature were observed in older rats. Higher levels of glycosaminoglycans were identified at PDL-bone and PDL-cementum entheses using alcian blue stain. Decreasing biochemical gradients from coronal to apical zones, specifically biomolecules that can induce osteogenic (biglycan) and fibrogenic (fibromodulin, decorin) phenotypes, and PDL-specific negative regulator of mineralization (asporin) were observed using immunohistochemistry. Heterogeneous distribution of Ca and P in alveolar bone, and relatively lower contents at the entheses, were observed using energy dispersive X-ray analysis. No correlation between age and microhardness of alveolar bone (0.7 ± 0.1 to 0.9 ± 0.2 GPa) and cementum (0.6 ± 0.1 to 0.8 ± 0.3 GPa) was observed using a microindenter. However, hardness of cementum and alveolar bone at any given age were significantly different (P<0.05). These observations should be taken into account as baseline parameters, during development (1.5 to 4 months), growth (4 to 10 months), followed by a senescent phase (10 to 15 months), from which deviations due to experimentally induced perturbations can be effectively investigated.
Collapse
Affiliation(s)
- Narita L. Leong
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| | - Jonathan M. Hurng
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| | - Sabra I. Djomehri
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| | - Stuart A. Gansky
- Division of Oral Epidemiology & Dental Public Health, Department of Preventive and Restorative Dental Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Mark I. Ryder
- Division of Periodontology, Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Sunita P. Ho
- Division of Biomaterials & Bioengineering, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
222
|
Hamamura K, Swarnkar G, Tanjung N, Cho E, Li J, Na S, Yokota H. RhoA-mediated signaling in mechanotransduction of osteoblasts. Connect Tissue Res 2012; 53:398-406. [PMID: 22420753 DOI: 10.3109/03008207.2012.671398] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Osteoblasts play a pivotal role in load-driven bone formation by activating Wnt signaling through a signal from osteocytes as a mechanosensor. Osteoblasts are also sensitive to mechanical stimulation, but the role of RhoA, a small GTPase involved in the regulation of cytoskeleton adhesion complexes, in mechanotransduction of osteoblasts is not completely understood. Using MC3T3-E1 osteoblast-like cells under 1 hr flow treatment at 10 dyn/cm(2), we examined a hypothesis that RhoA signaling mediates the cellular responses to flow-induced shear stress. To test the hypothesis, we conducted genome-wide pathway analysis and evaluated the role of RhoA in molecular signaling. Activity of RhoA was determined with a RhoA biosensor, which determined the activation state of RhoA based on a fluorescence resonance energy transfer between CFP and YFP fluorophores. A pathway analysis indicated that flow treatment activated phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling as well as a circadian regulatory pathway. Western blot analysis revealed that in response to flow treatment phosphorylation of Akt in PI3K signaling and phosphorylation of p38 and ERK1/2 in MAPK signaling were induced. FRET measurement showed that RhoA was activated by flow treatment, and an inhibitor to a Rho kinase significantly reduced flow-induced phosphorylation of p38, ERK1/2, and Akt as well as flow-driven elevation of the mRNA levels of osteopontin and cyclooxygenase-2. Collectively, the result demonstrates that in response to 1 hr flow treatment to MC3T3-E1 cells at 10 dyn/cm(2), RhoA plays a critical role in activating PI3K and MAPK signaling as well as modulating the circadian regulatory pathway.
Collapse
Affiliation(s)
- Kazunori Hamamura
- Center for Craniofacial Molecular Biology, University of Southern California School of Dentistry, Los Angeles, CA, USA
| | | | | | | | | | | | | |
Collapse
|
223
|
Miron RJ, Gruber R, Hedbom E, Saulacic N, Zhang Y, Sculean A, Bosshardt DD, Buser D. Impact of bone harvesting techniques on cell viability and the release of growth factors of autografts. Clin Implant Dent Relat Res 2012; 15:481-9. [PMID: 22375920 DOI: 10.1111/j.1708-8208.2012.00440.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Autogenous bone grafts obtained by different harvesting techniques behave differently during the process of graft consolidation; the underlying reasons are however not fully understood. One theory is that harvesting techniques have an impact on the number and activity of the transplanted cells which contribute to the process of graft consolidation. MATERIALS AND METHODS To test this assumption, porcine bone grafts were harvested with four different surgical procedures: bone mill, piezosurgery, bone drilling (bone slurry), and bone scraper. After determining cell viability, the release of molecules affecting bone formation and resorption was assessed by reverse transcription polymerase chain reaction and immunoassay. The mitogenic and osteogenic activity of the conditioned media was evaluated in a bioassay with isolated bone cells. RESULTS Cell viability and the release of molecules affecting bone formation were higher in samples harvested by bone mill and bone scraper when compared with samples prepared by bone drilling and piezosurgery. The harvesting procedure also affected gene expression, for example, bone mill and bone scraper samples revealed significantly higher expression of growth factors such as bone morphogenetic protein-2 and vascular endothelial growth factor compared with the two other modalities. Receptor activator of nuclear factor kappa B ligand expression was lowest in bone scraper samples. CONCLUSION These data can provide a scientific basis to better understand the impact of harvesting techniques on the number and activity of transplanted cells, which might contribute to the therapeutic outcome of the augmentation procedure.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Bern, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
224
|
Abstract
Bone remodeling and cartilage maintenance are strongly influenced by biomechanical signals generated by mechanical loading. Although moderate loading is required to maintain bone mass and cartilage homeostasis, loading can cause deleterious effects such as bone fracture and cartilage degradation. Because a tight coupling exists between cartilage and bone, alterations in one tissue can affect the other. Bone marrow lesions are often associated with an increased risk of developing cartilage defects, and changes in the articular cartilage integrity are linked to remodeling responses in the underlying bone. Although mechanisms regulating the maintenance of these two tissues are different, compelling evidence indicates that the signal pathways crosstalk, particularly with the Wnt pathway. A better understanding of the complex tempero-spatial interplay between bone remodeling and cartilage degeneration will help develop a therapeutic loading strategy that prevents bone loss and cartilage degeneration.
Collapse
Affiliation(s)
- Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
225
|
Abstract
Vertebrates evolved elaborating a structure made up of more than 200 bones and cartilages articulated with one another to form the skeleton, through which locomotion, organ protection, lodging of hematopoiesis, and mineral homeostasis are allowed. Skeletogenesis starts at the fetal stage, along with marrow hematopoiesis, and evolves postnatally through modeling and remodeling processes that permit skeletal mass buildup. Preservation of skeletal mass is then implemented by balanced remodeling, which ensures continuous renovation of the tissue to allow its mechanical, structural, and metabolic properties to remain unaltered until ageing or diseases disrupt this equilibrium. Skeletal homeostasis is fulfilled by specialized bone cells in association with systemic and local regulators. Herein I review landmark discoveries that shed light on the intricate mesh connecting bone cells among themselves and with other systems, thus representing the cellular basis of normal and abnormal bone development and homeostasis.
Collapse
Affiliation(s)
- Anna Teti
- Department of Experimental Medicine, University of L'Aquila, Via Vetoio-Coppito 2, 67100, L'Aquila, Italy.
| |
Collapse
|
226
|
Haley S, O'Grady S, Gulliver K, Bowman B, Baldassarre R, Miller S, Lane RH, Moyer-Mileur LJ. Mechanical-tactile stimulation (MTS) intervention in a neonatal stress model improves long-term outcomes on bone. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2011; 11:234-242. [PMID: 21885898 PMCID: PMC3228307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
OBJECTIVES Neonatal stress impairs postnatal bone mineralization. Evidence suggests that mechanical tactile stimulation (MTS) in early life decreases stress hormones and improves bone mineralization. Insulin-like growth factor (IGF1) is impacted by stress and essential to bone development. We hypothesized that MTS administered during neonatal stress would improve bone phenotype in later life. We also predicted an increase in bone specific mRNA expression of IGF1 related pathways. METHODS Neonatal stress (STRESS) and MTS (STRESS+10 min of MTS) were given from D6 to D10 of rat life and tissue was harvested on D60 of life. Dual energy x-ray absorptiometry (DXA), bone morphometry, serum osteocalcin, type I procollagen N-terminal propeptide (PINP), tartrate-resistant acid phosphatase (TRAP), and bone and liver mRNA levels of IGF1, IGF1 receptor (IGF1R), and growth hormone receptor (GHR) were measured. RESULTS Stress resulted in reduced bone area and bone mineral content (BMC) compared to naive control (CTL). MTS intervention increased BMC and tibial growth plate width compared to STRESS. MTS also resulted in higher osteocalcin, and, in males, lower TRAP (p<0.05). MTS resulted in three-fold, two-fold, and six-fold higher bone specific IGF1, IGF1R, and GHR, respectively (p ≤ 0.001) compared to STRESS. CONCLUSIONS MTS in early postnatal life improves long-term bone mineralization. IGF1 and related pathways may explain improved BMC.
Collapse
Affiliation(s)
- S Haley
- Center for Pediatric Nutrition Research, University of Utah, Salt Lake City, Utah
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Niver EL, Leong N, Greene J, Curtis D, Ryder MI, Ho SP. Reduced functional loads alter the physical characteristics of the bone-periodontal ligament-cementum complex. J Periodontal Res 2011; 46:730-41. [PMID: 21848615 DOI: 10.1111/j.1600-0765.2011.01396.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Adaptive properties of the bone-periodontal ligament-tooth complex have been identified by changing the magnitude of functional loads using small-scale animal models, such as rodents. Reported adaptive responses as a result of lower loads due to softer diet include decreased muscle development, change in structure-function relationship of the cranium, narrowed periodontal ligament space, and changes in the mineral level of the cortical bone and alveolar jaw bone and in the glycosaminoglycans of the alveolar bone. However, the adaptive role of the dynamic bone-periodontal ligament-cementum complex to prolonged reduced loads has not been fully explained to date, especially with regard to concurrent adaptations of bone, periodontal ligament and cementum. Therefore, in the present study, using a rat model, the temporal effect of reduced functional loads on physical characteristics, such as morphology and mechanical properties and the mineral profiles of the bone-periodontal ligament-cementum complex was investigated. MATERIAL AND METHODS Two groups of 6-wk-old male Sprague-Dawley rats were fed nutritionally identical food with a stiffness range of 127-158 N/mm for hard pellet or 0.3-0.5 N/mm for soft powder forms. Spatio-temporal adaptation of the bone-periodontal ligament-cementum complex was identified by mapping changes in the following: (i) periodontal ligament collagen orientation and birefringence using polarized light microscopy, bone and cementum adaptation using histochemistry, and bone and cementum morphology using micro-X-ray computed tomography; (ii) mineral profiles of the periodontal ligament-cementum and periodontal ligament-bone interfaces by X-ray attenuation; and (iii) microhardness of bone and cementum by microindentation of specimens at ages 6, 8, 12 and 15 wk. RESULTS Reduced functional loads over prolonged time resulted in the following adaptations: (i) altered periodontal ligament orientation and decreased periodontal ligament collagen birefringence, indicating decreased periodontal ligament turnover rate and decreased apical cementum resorption; (ii) a gradual increase in X-ray attenuation, owing to mineral differences, at the periodontal ligament-bone and periodontal ligament-cementum interfaces, without significant differences in the gradients for either group; (iii) significantly (p < 0.05) lower microhardness of alveolar bone (0.93 ± 0.16 GPa) and secondary cementum (0.803 ± 0.13 GPa) compared with the higher load group insert bone = (1.10 ± 0.17 and cementum = 0.940 ± 0.15 GPa, respectively) at 15 wk, indicating a temporal effect of loads on the local mineralization of bone and cementum. CONCLUSION Based on the results from this study, the effect of reduced functional loads for a prolonged time could differentially affect morphology, mechanical properties and mineral variations of the local load-bearing sites in the bone-periodontal ligament-cementum complex. These observed local changes in turn could help to explain the overall biomechanical function and adaptations of the tooth-bone joint. From a clinical translation perspective, our study provides an insight into modulation of load on the complex for improved tooth function during periodontal disease and/or orthodontic and prosthodontic treatments.
Collapse
Affiliation(s)
- E L Niver
- Division of Periodontology, Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
228
|
Boccafoschi F, Mosca C, Bosetti M, Cannas M. The role of mechanical stretching in the activation and localization of adhesion proteins and related intracellular molecules. J Cell Biochem 2011; 112:1403-9. [PMID: 21321993 DOI: 10.1002/jcb.23056] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The molecular complexity of the processes which lead to cell adhesion includes membrane and cytoskeletal proteins, involved in the focal adhesion formation, as well as signaling molecules tightly associated with the main intracellular regulatory cascades (Akt/PKB and MAPK/Erk). Dynamic environments, which create substrate deformations at determined frequencies and timing, have significant influences on adhesion mechanisms and in general in cellular behavior. In this work, we investigated the role of mechanical stretching (10% substrate deformation, 1 Hz frequency applied up to 60 min) on adhesion proteins (vinculin and focal adhesion kinase-FAK), related RhoGTPases (Rac1 and RhoA), and intracellular pathways (Akt/PKB and MAPK/Erk) in terms of activation and membrane recruitment in relation with cytoskeletal changes observed (membrane ruffling and filopodia formation). These changes are due to intracellular molecular rearrangements, acting with sequential concerted dynamics, able to modify the cytoskeletal conformation. The observed cellular response adds some important issues for better understanding the cellular behavior in environment which mimic as close as possible the physiological conditions.
Collapse
Affiliation(s)
- F Boccafoschi
- Department of Clinical and Experimental Medicine, University of Piemonte Orientale "A. Avogadro", 28100 Novara, Italy.
| | | | | | | |
Collapse
|
229
|
Hurng JM, Kurylo MP, Marshall GW, Webb SM, Ryder MI, Ho SP. Discontinuities in the human bone-PDL-cementum complex. Biomaterials 2011; 32:7106-17. [PMID: 21774982 DOI: 10.1016/j.biomaterials.2011.06.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 06/09/2011] [Indexed: 12/20/2022]
Abstract
A naturally graded interface due to functional demands can deviate toward a discontinuous interface, eventually decreasing the functional efficiency of a dynamic joint. It is this characteristic feature in a human bone-tooth fibrous joint bone-PDL-tooth complex that will be discussed through histochemistry, and site-specific high resolution microscopy, micro tomography(Micro XCT™), X-ray fluorescence imaging and wet nanoindentation techniques. Results demonstrated two causes for the occurrence of 5-50 μm narrowed PDL-space: 1) microscopic scalloped regions at the PDL-insertion sites and macro-scale stratified layers of bone with rich basophilic lines, and 2) macroscopic bony protrusions. Narrowed PDL-complexes illustrated patchy appearance of asporin, and when imaged under wet conditions using an atomic force microscope (AFM), demonstrated structural reorganization of the PDL, collagen periodicity, organic-dominant areas at the PDL-cementum and PDL-bone entheses and within cementum and bone. Scanning electron microscopy (SEM) results confirmed AFM results. Despite the narrowed PDL, continuity between PDL and vasculature in endosteal spaces of bone was demonstrated using a Micro XCT™. The higher levels of Ca and P X-ray fluorescence using a microprobe were correlated with higher elastic modulus values of 0.1-1.4 and 0.1-1.2 GPa for PDL-bone and PDL-cementum using wet nanoindentation. The ranges in elastic modulus values for PDL-bone and PDL-cementum entheses in 150-380 μm wide PDL-complex were 0.1-1.0 and 0.1-0.6 GPa. Based on these results we propose that strain amplification at the entheses could be minimized with a gradual change in modulus profile, a characteristic of 150-380 μm wide functional PDL-space. However, a discontinuity in modulus profile, a characteristic of 5-50 μm wide narrowed PDL-space would cause compromised mechanotransduction. The constrictions or narrowed sites within the bone-tooth fibrous joint will become the new "load bearing sites" that eventually could cause direct local fusion of bone with cementum.
Collapse
Affiliation(s)
- Jonathan M Hurng
- Division of Biomaterials and Bioengineering, Department of Preventive and Restorative Dental Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
230
|
Chen LL, Lei LH, Ding PH, Tang Q, Wu YM. Osteogenic effect of Drynariae rhizoma extracts and Naringin on MC3T3-E1 cells and an induced rat alveolar bone resorption model. Arch Oral Biol 2011; 56:1655-62. [PMID: 21764032 DOI: 10.1016/j.archoralbio.2011.06.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 05/27/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To investigate if Drynariae rhizoma (DR) and its main ingredient Naringin could reduce alveolar bone loss by stimulating the proliferation and differentiation of osteoblasts. MATERIALS AND METHODS The effect of DR water (DRWE), ethanolic extract (DREE), and Naringin on MC3T3-E1 cells was evaluated respectively by MTT method and by measuring the activity of alkaline phosphatase (ALP activity) as well as the level of osteocalcin in medium. Bone mineral density (BMD) detection, osteoclast counting by tartrate resistant acid phosphatase staining, and histopathological analysis were performed in an induced rat model of alveolar bone resorption after gastric perfusion with DR extracts or Naringin. RESULTS DRWE and Naringin effectively increased the proliferation of MC3T3-E1 cells, whilst DREE and Naringin enhanced the differentiation of osteoblastic cells. The in vivo study indicated an elevated BMD value in the tooth-periodontal tissues from DRWE, DREE and Naringin treated groups after 10, 20 and 30 days of perfusion (P<0.05). In DRWE treated group, the number of osteoclasts at days 10, 20 and 30 decreased remarkably as compared to the corresponding negative controls (P<0.05), and no osteoclast could be found at day 30. New non-calcified bone-like matrix attached by osteoblasts at the root furcation was also shown. CONCLUSIONS DR could be a supplementary medicine for periodontal therapy as it could reduce bone resorption in rat model of alveolar bone resorption and exert osteogenic effect on osteoblasts.
Collapse
Affiliation(s)
- Li-Li Chen
- Department of Oral Medicine, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | | | | | | | | |
Collapse
|
231
|
Peng Q, Wang Y, Qiu J, Zhang B, Sun J, Lv Y, Yang L. A novel mechanical loading model for studying the distributions of strain and mechano-growth factor expression. Arch Biochem Biophys 2011; 511:8-13. [DOI: 10.1016/j.abb.2011.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 04/19/2011] [Accepted: 04/22/2011] [Indexed: 01/06/2023]
|
232
|
Totland GK, Fjelldal PG, Kryvi H, Løkka G, Wargelius A, Sagstad A, Hansen T, Grotmol S. Sustained swimming increases the mineral content and osteocyte density of salmon vertebral bone. J Anat 2011; 219:490-501. [PMID: 21615400 DOI: 10.1111/j.1469-7580.2011.01399.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study addresses the effects of increased mechanical load on the vertebral bone of post-smolt Atlantic salmon by forcing them to swim at controlled speeds. The fish swam continuously in four circular tanks for 9 weeks, two groups at 0.47 body lengths (bl) × s(-1) (non-exercised group) and two groups at 2 bl × s(-1) (exercised group), which is just below the limit for maximum sustained swimming speed in this species. Qualitative data concerning the vertebral structure were obtained from histology and electron microscopy, and quantitative data were based on histomorphometry, high-resolution X-ray micro-computed tomography images and analysis of bone mineral content, while the mechanical properties were tested by compression. Our key findings are that the bone matrix secreted during sustained swimming had significantly higher mineral content and mechanical strength, while no effect was detected on bone in vivo architecture. mRNA levels for two mineralization-related genes bgp and alp were significantly upregulated in the exercised fish, indicating promotion of mineralization. The osteocyte density of the lamellar bone of the amphicoel was also significantly higher in the exercised than non-exercised fish, while the osteocyte density in the cancellous bone was similar in the two groups. The vertebral osteocytes did not form a functional syncytium, which shows that salmon vertebral bone responds to mechanical loading in the absence of an extensive connecting syncytial network of osteocytic cell processes as found in mammals, indicating the existence of a different mechanosensing mechanism. The adaptive response to increased load is thus probably mediated by osteoblasts or bone lining cells, a system in which signal detection and response may be co-located. This study offers new insight into the teleost bone biology, and may have implications for maintaining acceptable welfare for farmed salmon.
Collapse
|
233
|
Serrat MA, Williams RM, Farnum CE. Exercise mitigates the stunting effect of cold temperature on limb elongation in mice by increasing solute delivery to the growth plate. J Appl Physiol (1985) 2010; 109:1869-79. [PMID: 20930127 DOI: 10.1152/japplphysiol.01022.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ambient temperature and physical activity modulate bone elongation in mammals, but mechanisms underlying this plasticity are a century-old enigma. Longitudinal bone growth occurs in cartilaginous plates, which receive nutritional support via delivery of solutes from the vasculature. We tested the hypothesis that chronic exercise and warm temperature promote bone lengthening by increasing solute delivery to the growth plate, measured in real time using in vivo multiphoton microscopy. We housed 68 weanling female mice at cold (16°C) or warm (25°C) temperatures and allowed some groups voluntary access to a running wheel. We show that exercise mitigates the stunting effect of cold temperature on limb elongation after 11 days of wheel running. All runners had significantly lengthened limbs, regardless of temperature, while nonrunning mice had shorter limbs that correlated with housing temperature. Tail length was impacted only by temperature, indicating that the exercise effect was localized to limb bones and was not a systemic endocrine reaction. In vivo multiphoton imaging of fluoresceinated tracers revealed enhanced solute delivery to tibial growth plates in wheel-running mice, measured under anesthesia at rest. There was a minimal effect of rearing temperature on solute delivery when measured at an intermediate room temperature (20°C), suggesting that a lasting increase in solute delivery is an important factor in exercise-mediated limb lengthening but may not play a role in temperature-mediated limb lengthening. These results are relevant to the study of skeletal evolution in mammals from varying environments and have the potential to fundamentally advance our understanding of bone elongation processes.
Collapse
Affiliation(s)
- Maria A Serrat
- Department of Anatomy and Pathology, Joan C. Edwards School of Medicine, Marshall University, 1542 Spring Valley Dr., Huntington, WV 25704, USA.
| | | | | |
Collapse
|
234
|
Bellido T. Antagonistic interplay between mechanical forces and glucocorticoids in bone: a tale of kinases. J Cell Biochem 2010; 111:1-6. [PMID: 20506204 PMCID: PMC10132774 DOI: 10.1002/jcb.22660] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The contrasting actions of mechanical forces and glucocorticoids (GC) on bone have been long recognized. However, the cellular and molecular mechanisms by which these stimuli impact the skeleton remain only partially known. Recent evidence gained from studies on bone cell apoptosis has revealed that mechanical forces and GC exhibit converse effects on osteocyte and osteoblast survival resulting from divergent actions on the focal adhesion kinases FAK and Pyk2, molecules that regulate integrin-dependent interactions between bone cells and the extracellular matrix (ECM). This prospect reviews these findings and poses the possibility that similar opposing effects on kinase signaling are responsible for other actions of mechanical forces and GC on the skeleton, in particular on bone formation and the Wnt signaling pathway.
Collapse
Affiliation(s)
- Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
235
|
McAteer ME, Niziolek PJ, Ellis SN, Alge DL, Robling AG. Mechanical stimulation and intermittent parathyroid hormone treatment induce disproportional osteogenic, geometric, and biomechanical effects in growing mouse bone. Calcif Tissue Int 2010; 86:389-96. [PMID: 20306026 PMCID: PMC3412136 DOI: 10.1007/s00223-010-9348-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 02/26/2010] [Indexed: 11/25/2022]
Abstract
Mechanical loading and intermittent parathyroid (iPTH) treatment are both osteoanabolic stimuli and are regulated by partially overlapping cellular signaling pathways. iPTH has been shown clinically to be effective in increasing bone mass and reducing fracture risk. Likewise, mechanical stimulation can significantly enhance bone apposition and prevent bone loss, but its clinical effects on fracture susceptibility are less certain. Many of the osteogenic effects of iPTH are localized to biomechanically suboptimal bone surfaces, whereas mechanical loading directs new bone formation to high-stress areas and not to strain-neutral areas. These differences in localization in new tissue, resulting from load-induced versus iPTH-induced bone accumulation, should affect the relation between bone mass and bone strength, or "tissue economy." We investigated the changes in bone mass and strength induced by 6 weeks of mechanical loading and compared them to changes induced by 6 weeks of iPTH treatment. Loading and iPTH both increased ulnar bone accrual, as measured by bone mineral density and content, and fluorochrome-derived bone formation. iPTH induced a significantly greater increase in bone mass than loading, but ulnar bone strength was increased approximately the same amount by both treatments. Mechanical loading during growth can spatially optimize new bone formation to improve structural integrity with a minimal increase in mass, thereby increasing tissue economy, i.e., the amount of strength returned per unit bone mass added. Furthermore, exercise studies in which only small changes in bone mass are detected might be more beneficial to bone health and fracture resistance than has commonly been presumed.
Collapse
Affiliation(s)
- Maureen E. McAteer
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul J. Niziolek
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Shana N. Ellis
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daniel L. Alge
- Department of Biomedical Engineering, Indiana University–Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Alexander G. Robling
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Engineering, Indiana University–Purdue University at Indianapolis, Indianapolis, IN, USA
| |
Collapse
|