1
|
Wherry SJ, Miller RM, Jeong SH, Beavers KM. The Ability of Exercise to Mitigate Caloric Restriction-Induced Bone Loss in Older Adults: A Structured Review of RCTs and Narrative Review of Exercise-Induced Changes in Bone Biomarkers. Nutrients 2021; 13:1250. [PMID: 33920153 PMCID: PMC8070587 DOI: 10.3390/nu13041250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 11/17/2022] Open
Abstract
Despite the adverse metabolic and functional consequences of obesity, caloric restriction- (CR) induced weight loss is often contra-indicated in older adults with obesity due to the accompanying loss of areal bone mineral density (aBMD) and subsequent increased risk of fracture. Several studies show a positive effect of exercise on aBMD among weight-stable older adults; however, data on the ability of exercise to mitigate bone loss secondary to CR are surprisingly equivocal. The purpose of this review is to provide a focused update of the randomized controlled trial literature assessing the efficacy of exercise as a countermeasure to CR-induced bone loss among older adults. Secondarily, we present data demonstrating the occurrence of exercise-induced changes in bone biomarkers, offering insight into why exercise is not more effective than observed in mitigating CR-induced bone loss.
Collapse
Affiliation(s)
- Sarah J. Wherry
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
- VA Eastern Colorado Geriatric Research, Education, and Clinical Center (GRECC), Aurora, CO 80045, USA
| | - Ryan M. Miller
- Department of Internal Medicine, Sections on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
| | - Sarah H. Jeong
- Z. Smith Reynolds Library, Wake Forest University, Winston-Salem, NC 27109, USA;
| | - Kristen M. Beavers
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, NC 27109, USA
| |
Collapse
|
2
|
Chermside-Scabbo CJ, Harris TL, Brodt MD, Braenne I, Zhang B, Farber CR, Silva MJ. Old Mice Have Less Transcriptional Activation But Similar Periosteal Cell Proliferation Compared to Young-Adult Mice in Response to in vivo Mechanical Loading. J Bone Miner Res 2020; 35:1751-1764. [PMID: 32311160 PMCID: PMC7486279 DOI: 10.1002/jbmr.4031] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Mechanical loading is a potent strategy to induce bone formation, but with aging, the bone formation response to the same mechanical stimulus diminishes. Our main objectives were to (i) discover the potential transcriptional differences and (ii) compare the periosteal cell proliferation between tibias of young-adult and old mice in response to strain-matched mechanical loading. First, to discover potential age-related transcriptional differences, we performed RNA sequencing (RNA-seq) to compare the loading responses between tibias of young-adult (5-month) and old (22-month) C57BL/6N female mice following 1, 3, or 5 days of axial loading (loaded versus non-loaded). Compared to young-adult mice, old mice had less transcriptional activation following loading at each time point, as measured by the number of differentially expressed genes (DEGs) and the fold-changes of the DEGs. Old mice engaged fewer pathways and gene ontology (GO) processes, showing less activation of processes related to proliferation and differentiation. In tibias of young-adult mice, we observed prominent Wnt signaling, extracellular matrix (ECM), and neuronal responses, which were diminished with aging. Additionally, we identified several targets that may be effective in restoring the mechanoresponsiveness of aged bone, including nerve growth factor (NGF), Notum, prostaglandin signaling, Nell-1, and the AP-1 family. Second, to directly test the extent to which periosteal cell proliferation was diminished in old mice, we used bromodeoxyuridine (BrdU) in a separate cohort of mice to label cells that divided during the 5-day loading interval. Young-adult and old mice had an average of 15.5 and 16.7 BrdU+ surface cells/mm, respectively, suggesting that impaired proliferation in the first 5 days of loading does not explain the diminished bone formation response with aging. We conclude that old mice have diminished transcriptional activation following mechanical loading, but periosteal proliferation in the first 5 days of loading does not differ between tibias of young-adult and old mice. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christopher J Chermside-Scabbo
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Medical Scientist Training Program, Washington University School of Medicine, Washington University, St. Louis, MO, USA
| | - Taylor L Harris
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Michael D Brodt
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
| | - Ingrid Braenne
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Department of Developmental Biology, Washington University, St. Louis, MO, USA
| | - Charles R Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Matthew J Silva
- Musculoskeletal Research Center Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| |
Collapse
|
3
|
Abstract
Prostaglandins (PGs) are highly bioactive fatty acids. PGs, especially prostaglandin E2 (PGE2), are abundantly produced by cells of both the bone-forming (osteoblast) lineage and the bone-resorbing (osteoclast) lineage. The inducible cyclooxygenase, COX-2, is largely responsible for most PGE2 production in bone, and once released, PGE2 is rapidly degraded in vivo. COX-2 is induced by multiple agonists - hormones, growth factors, and proinflammatory factors - and the resulting PGE2 may mediate, amplify, or, as we have recently shown for parathyroid hormone (PTH), inhibit responses to these agonists. In vitro, PGE2 can directly stimulate osteoblast differentiation and, indirectly via stimulation of RANKL in osteoblastic cells, stimulate the differentiation of osteoclasts. The net balance of these two effects of PGE2 in vivo on bone formation and bone resorption has been hard to predict and, as expected for such a widespread local factor, hard to study. Some of the complexity of PGE2 actions on bone can be explained by the fact that there are four receptors for PGE2 (EP1-4). Some of the major actions of PGE2 in vitro occur via EP2 and EP4, both of which can stimulate cAMP signaling, but there are other distinct signaling pathways, important in other tissues, which have not yet been fully elucidated in bone cells. Giving PGE2 or agonists of EP2 and EP4 to accelerate bone repair has been examined with positive results. Further studies to clarify the pathways of PGE2 action in bone may allow us to identify new and more effective ways to deliver the therapeutic benefits of PGE2 in skeletal disorders.
Collapse
Affiliation(s)
- Carol Pilbeam
- Department of Medicine and Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| |
Collapse
|
4
|
Morse A, Schindeler A, McDonald MM, Kneissel M, Kramer I, Little DG. Sclerostin Antibody Augments the Anabolic Bone Formation Response in a Mouse Model of Mechanical Tibial Loading. J Bone Miner Res 2018; 33:486-498. [PMID: 29090474 DOI: 10.1002/jbmr.3330] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/11/2017] [Accepted: 10/29/2017] [Indexed: 12/18/2022]
Abstract
Decreased activity or expression of sclerostin, an endogenous inhibitor of Wnt/β-catenin signaling, results in increased bone formation and mass. Antibodies targeting and neutralizing sclerostin (Scl-Ab) have been shown to increase bone mass and reduce fracture risk. Sclerostin is also important in modulating the response of bone to changes in its biomechanical environment. However, the effects of Scl-Ab on mechanotransduction are unclear, and it was speculated that the loading response may be altered for individuals receiving Scl-Ab therapy. To address this, we carried out a 2-week study of tibial cyclic compressive loading on C57Bl/6 mice treated with vehicle or 100 mg/kg/wk Scl-Ab. Increases in bone volume, density, and dynamic bone formation were found with loading, and the anabolic response was further increased by the combination of load and Scl-Ab. To investigate the underlying mechanism, gene profiling by RNA sequencing (RNAseq) was performed on tibias isolated from mice from all four experimental groups. Major alterations in Wnt/β-catenin gene expression were found with tibial loading, however not with Scl-Ab treatment alone. Notably, the combination of load and Scl-Ab elicited a synergistic response from a number of specific Wnt-related and mechanotransduction factors. An unexpected finding was significant upregulation of factors in the Rho GTPase signaling pathway with combination treatment. In summary, combination therapy had a more profound anabolic response than either Scl-Ab or loading treatment alone. The Wnt/β-catenin and Rho GTPase pathways were implicated within bone mechanotransduction and support the concept that bone mechanotransduction is likely to encompass a number of interconnected signaling pathways. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Michelle M McDonald
- Bone Biology Program, The Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
5
|
Laurent MR, Dubois V, Claessens F, Verschueren SMP, Vanderschueren D, Gielen E, Jardí F. Muscle-bone interactions: From experimental models to the clinic? A critical update. Mol Cell Endocrinol 2016; 432:14-36. [PMID: 26506009 DOI: 10.1016/j.mce.2015.10.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
Bone is a biomechanical tissue shaped by forces from muscles and gravitation. Simultaneous bone and muscle decay and dysfunction (osteosarcopenia or sarco-osteoporosis) is seen in ageing, numerous clinical situations including after stroke or paralysis, in neuromuscular dystrophies, glucocorticoid excess, or in association with vitamin D, growth hormone/insulin like growth factor or sex steroid deficiency, as well as in spaceflight. Physical exercise may be beneficial in these situations, but further work is still needed to translate acceptable and effective biomechanical interventions like vibration therapy from animal models to humans. Novel antiresorptive and anabolic therapies are emerging for osteoporosis as well as drugs for sarcopenia, cancer cachexia or muscle wasting disorders, including antibodies against myostatin or activin receptor type IIA and IIB (e.g. bimagrumab). Ideally, increasing muscle mass would increase muscle strength and restore bone loss from disuse. However, the classical view that muscle is unidirectionally dominant over bone via mechanical loading is overly simplistic. Indeed, recent studies indicate a role for neuronal regulation of not only muscle but also bone metabolism, bone signaling pathways like receptor activator of nuclear factor kappa-B ligand (RANKL) implicated in muscle biology, myokines affecting bone and possible bone-to-muscle communication. Moreover, pharmacological strategies inducing isolated myocyte hypertrophy may not translate into increased muscle power because tendons, connective tissue, neurons and energy metabolism need to adapt as well. We aim here to critically review key musculoskeletal molecular pathways involved in mechanoregulation and their effect on the bone-muscle unit as a whole, as well as preclinical and emerging clinical evidence regarding the effects of sarcopenia therapies on osteoporosis and vice versa.
Collapse
Affiliation(s)
- Michaël R Laurent
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium.
| | - Vanessa Dubois
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Sabine M P Verschueren
- Research Group for Musculoskeletal Rehabilitation, Department of Rehabilitation Science, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Evelien Gielen
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
6
|
Jankowski CM, Shea K, Barry DW, Linnebur SA, Wolfe P, Kittelson J, Schwartz RS, Kohrt WM. Timing of Ibuprofen Use and Musculoskeletal Adaptations to Exercise Training in Older Adults. Bone Rep 2015; 1:1-8. [PMID: 25642444 PMCID: PMC4310009 DOI: 10.1016/j.bonr.2014.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Prostaglandins (PGs) increase in bone in response to mechanical loading and stimulate bone formation. Inhibition of cyclooxygenase (COX), the enzyme responsible for PG synthesis, by non-steroidal anti-inflammatory drugs (NSAIDs) impairs the bone formation response to loading in animals when administered before, but not after, loading. The aim was to determine whether the timing of ibuprofen use (400 mg before versus after exercise sessions) is a significant determinant of the adaptive response of BMD to exercise training in older adults. We hypothesized that taking ibuprofen before exercise would attenuate the improvements in total hip and lumbar spine BMD in response to 36 weeks of training when compared with placebo or with ibuprofen use after exercise. Untrained women and men (N = 189) aged 60 to 75 years were randomly assigned to 1 of 3 treatment arms: placebo before and after exercise (PP); ibuprofen before and placebo after exercise (IP); and placebo before and ibuprofen after exercise (PI). The difference between groups in the change in BMD was not significant when IP was compared with either PP (hip, − 0.5% (− 1.4, 0.4); spine, 0.1% (− 0.9, 1.2)) or PI (hip, 0.3% (− 0.6, 1.2); spine, 0.5% (− 0.5, 1.5)). Ibuprofen use appeared to have more adverse effects on BMD in women than men. The study demonstrated that ibuprofen use did not significantly alter the BMD adaptations to exercise in older adults, but this finding should be interpreted cautiously. It had been expected that the inhibition of bone formation by ibuprofen would be more robust in men than in women, but this did not appear to be the case and may have limited the power to detect the effects of ibuprofen. Further research is needed to understand whether NSAID use counteracts, in part, the beneficial effects of exercise on bone. The purpose was to determine whether musculoskeletal adaptations to exercise training in older adults are influenced by NSAID use. Ibuprofen use did not significantly alter changes in BMD or fat-free mass, but the study may have been inadequately powered. Study attrition was significantly lower in the group that took NSAIDs before exercise suggesting improved tolerance of vigorous bone-loading exercise. This was the first randomized, double-blinded placebo- controlled study of ibuprofen effects on BMD from exercise in an older population.
Collapse
Affiliation(s)
- Catherine M. Jankowski
- College of Nursing, University of Colorado Anschutz Medical Campus, USA
- Department of Medicine, Division of Geriatric Medicine, University of Colorado, Anschutz Medical Campus, USA
- Corresponding author at: Mail Stop C288-19, 13120 East 19th Avenue, Aurora, CO 80045, USA.
| | - Karen Shea
- Department of Medicine, Division of Geriatric Medicine, University of Colorado, Anschutz Medical Campus, USA
| | - Daniel W. Barry
- Department of Medicine, General Internal Medicine, University of Colorado Anschutz Medical Campus, USA
| | - Sunny A. Linnebur
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, USA
| | - Pamela Wolfe
- Department of Preventive Medicine and Biometrics, University of Colorado Anschutz Medical Campus, USA
| | - John Kittelson
- Department of Preventive Medicine and Biometrics, University of Colorado Anschutz Medical Campus, USA
| | - Robert S. Schwartz
- Department of Medicine, Division of Geriatric Medicine, University of Colorado, Anschutz Medical Campus, USA
| | - Wendy M. Kohrt
- Department of Medicine, Division of Geriatric Medicine, University of Colorado, Anschutz Medical Campus, USA
| |
Collapse
|
7
|
Ruggiu A, Cancedda R. Bone mechanobiology, gravity and tissue engineering: effects and insights. J Tissue Eng Regen Med 2014; 9:1339-51. [PMID: 25052837 DOI: 10.1002/term.1942] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 05/23/2014] [Accepted: 05/27/2014] [Indexed: 01/10/2023]
Abstract
Bone homeostasis strongly depends on fine tuned mechanosensitive regulation signals from environmental forces into biochemical responses. Similar to the ageing process, during spaceflights an altered mechanotransduction occurs as a result of the effects of bone unloading, eventually leading to loss of functional tissue. Although spaceflights represent the best environment to investigate near-zero gravity effects, there are major limitations for setting up experimental analysis. A more feasible approach to analyse the effects of reduced mechanostimulation on the bone is represented by the 'simulated microgravity' experiments based on: (1) in vitro studies, involving cell cultures studies and the use of bioreactors with tissue engineering approaches; (2) in vivo studies, based on animal models; and (3) direct analysis on human beings, as in the case of the bed rest tests. At present, advanced tissue engineering methods allow investigators to recreate bone microenvironment in vitro for mechanobiology studies. This group and others have generated tissue 'organoids' to mimic in vitro the in vivo bone environment and to study the alteration cells can go through when subjected to unloading. Understanding the molecular mechanisms underlying the bone tissue response to mechanostimuli will help developing new strategies to prevent loss of tissue caused by altered mechanotransduction, as well as identifying new approaches for the treatment of diseases via drug testing. This review focuses on the effects of reduced gravity on bone mechanobiology by providing the up-to-date and state of the art on the available data by drawing a parallel with the suitable tissue engineering systems.
Collapse
Affiliation(s)
- Alessandra Ruggiu
- University of Genova, Department of Experimental Medicine, Genova, Italy
| | - Ranieri Cancedda
- University of Genova, Department of Experimental Medicine & IRCCS AOU San Martino-IST, National Institute for Cancer Research, Genova, Italy
| |
Collapse
|
8
|
Castillo AB, Triplett JW, Pavalko FM, Turner CH. Estrogen receptor-β regulates mechanical signaling in primary osteoblasts. Am J Physiol Endocrinol Metab 2014; 306:E937-44. [PMID: 24619882 PMCID: PMC3989741 DOI: 10.1152/ajpendo.00458.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanical loading is an important regulator in skeletal growth, maintenance, and aging. Estrogen receptors have a regulatory role in mechanically induced bone adaptation. Estrogen receptor-α (ERα) is known to enhance load-induced bone formation, whereas ERβ negatively regulates this process. We hypothesized that ERβ regulates mechanical signaling in osteoblasts. We tested this hypothesis by subjecting primary calvarial cells isolated from wild-type and ERβ-knockout mice (BERKO) to oscillatory fluid flow in the absence or presence of estradiol (E2). We found that the known responses to fluid shear stress, i.e., phosphorylation of the mitogen-activated protein kinase ERK and upregulation of COX-2 expression, were inhibited in BERKO cells in the absence of E2. Flow-induced increase in prostaglandin E2 (PGE2) release was not altered in BERKO cells in the absence of E2, but was increased when E2 was present. Additionally, immunofluorescence analysis and estrogen response element luciferase assays revealed increased ERα expression and flow- and ligand-induced nuclear translocation as well as transcriptional activity in BERKO cells in both the presence and absence of E2. Taken together, these data suggest that ERβ plays both ligand-dependent and ligand-independent roles in mechanical signaling in osteoblasts. Furthermore, our data suggest that one mechanism by which ERβ regulates mechanotransduction in osteoblasts may result from its inhibitory effect on ERα expression and function. Targeting estrogen receptors (e.g., inhibiting ERβ) may represent an effective approach for prevention and treatment of age-related bone loss.
Collapse
Affiliation(s)
- Alesha B Castillo
- Rehabilitation Research and Development, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | | | | | | |
Collapse
|
9
|
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
|
10
|
Bivi N, Pacheco-Costa R, Brun LR, Murphy TR, Farlow NR, Robling AG, Bellido T, Plotkin LI. Absence of Cx43 selectively from osteocytes enhances responsiveness to mechanical force in mice. J Orthop Res 2013; 31:1075-81. [PMID: 23483620 PMCID: PMC3663897 DOI: 10.1002/jor.22341] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 02/08/2013] [Indexed: 02/04/2023]
Abstract
The osteocyte network is crucial for the response of bone to mechanical force. Within this network, connexin43 (Cx43) is thought to mediate the communication of osteocytes and osteoblasts among themselves and the exchange of small molecules with the extracellular milieu. Despite recent advances in understanding Cx43 role for the response of bone cells to mechanical stimulation, the contribution of Cx43 specifically in osteocytes to mechanotransduction in vivo is not well-known. We examined the anabolic response to ulnar axial loading of mice lacking Cx43 in osteocytes (Cx43(ΔOt)). Loading induced a greater increase in periosteal bone formation rate in Cx43(ΔOt) mice compared to control littermates, resulting from higher mineralizing surface and enhanced mineral apposition rate. Expression of β-catenin protein, a molecule implicated in mechanotransduction, was higher in bones from Cx43(ΔOt) mice, compared to littermate controls. In addition, MLO-Y4 osteocytic cells knocked-down for Cx43 exhibited higher β-catenin protein expression and enhanced response to mechanical stimulation. These findings suggest that osteocytes lacking Cx43 are "primed" to respond to mechanical stimulation and that absence of Cx43 in osteocytes unleashes bone formation, by a mechanism that might involve accumulation of β-catenin.
Collapse
Affiliation(s)
- Nicoletta Bivi
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US
| | - Rafael Pacheco-Costa
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US,Department of Functional & Structural Biology, Federal University of São Paulo, São Paulo, Brazil
| | - Lucas R. Brun
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US
| | - Thomas R. Murphy
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US
| | - Nathan R. Farlow
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US
| | - Alexander G. Robling
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US
| | - Teresita Bellido
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US,Div. Endocrinology, Dept. Internal Medicine, Indiana University School of Medicine, Indianapolis, IN, US
| | - Lilian I. Plotkin
- Dept. Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, US,Corresponding author: Lilian I. Plotkin, Ph.D. Department of Anatomy and Cell Biology Indiana University School of Medicine 635 Barnhill Drive, MS-5035 Indianapolis, IN 46202-5120 Phone: 1-317-274-5317 Fax: 1-317-278-2040
| |
Collapse
|
11
|
Plotkin LI, Bellido T. Beyond gap junctions: Connexin43 and bone cell signaling. Bone 2013; 52:157-66. [PMID: 23041511 PMCID: PMC3513515 DOI: 10.1016/j.bone.2012.09.030] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 09/20/2012] [Accepted: 09/25/2012] [Indexed: 12/31/2022]
Abstract
Connexin43 (Cx43) is the most abundant gap junction protein expressed in bone cells and plays a central role in cell-to-cell communication in the skeleton. Findings of the last decade uncovered functions of Cx43 hemichannels expressed on unopposed plasma cell membranes as mediators of the communication between bone cells and their extracellular milieu. Additionally, through its cytoplasmic C-terminus domain, Cx43 serves as a scaffolding protein that associates with structural and signaling molecules leading to regulation of intracellular signaling, independent of channel activity. This perspective discusses the evidence demonstrating that via these diverse mechanisms Cx43 is a key component of the intracellular machinery responsible for signal transduction in bone in response to pharmacologic, hormonal and mechanical stimuli. This advance in the knowledge of the role of connexins increases our understanding of the pathophysiological mechanisms that regulate bone cell function and provides new opportunities to treat bone diseases.
Collapse
Affiliation(s)
- Lilian I. Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
12
|
Sugiyama T, Meakin LB, Galea GL, Lanyon LE, Price JS. The cyclooxygenase-2 selective inhibitor NS-398 does not influence trabecular or cortical bone gain resulting from repeated mechanical loading in female mice. Osteoporos Int 2013; 24:383-8. [PMID: 22349912 PMCID: PMC3536947 DOI: 10.1007/s00198-012-1922-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 01/17/2012] [Indexed: 11/30/2022]
Abstract
UNLABELLED A single injection of the cyclooxygenase-2 (COX-2) selective inhibitor NS-398 reduces bone's osteogenic response to a single period of mechanical loading in female rats, while women taking COX-2 selective inhibitors do not have lower bone mass. We show that daily NS-398 injection does not influence bone gain from repeated loading in female mice. INTRODUCTION Prostaglandins are mediators of bone cells' early response to mechanical stimulation. COX-2 expression is up-regulated by exposure of these cells to mechanical strain or fluid flow, and the osteogenic response to a single loading period is reduced by COX-2 inhibition. This study determined, in female mice in vivo, the effect of longer term COX-2 inhibition on adaptive (re)modelling of cortical and trabecular bone in response to repeated loading. METHODS Nineteen-week-old female C57BL/6 mice were injected with vehicle or NS-398 (5 mg/kg/day) 5 days a week for 2 weeks. On three alternate days each week, the right tibiae/fibulae were axially loaded [40 cycles (7 min)/day] three hours after injection. Left limbs acted as internal controls. Changes in three-dimensional bone architecture were analysed by high-resolution micro-computed tomography. RESULTS In control limbs NS-398 was associated with reduced trabecular number but had no influence on cortical bone. In loaded limbs trabecular thickness and cortical periosteally enclosed volume increased. NS-398 showed no effect on this response. CONCLUSION Pharmacological inhibition of COX-2 by NS-398 does not affect trabecular or cortical bone's response to repeated mechanical loading in female mice and thus would not be expected to impair the functional adaptation of bone to physical activity in women.
Collapse
Affiliation(s)
- T Sugiyama
- Department of Veterinary Basic Sciences, The Royal Veterinary College, University of London, London, UK.
| | | | | | | | | |
Collapse
|
13
|
Bao H, Ge Y, Zhuang S, Dworkin LD, Liu Z, Gong R. Inhibition of glycogen synthase kinase-3β prevents NSAID-induced acute kidney injury. Kidney Int 2012; 81:662-73. [PMID: 22258319 PMCID: PMC3305839 DOI: 10.1038/ki.2011.443] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Clinical use of nonsteroidal anti-inflammatory drugs (NSAIDs) like diclofenac (DCLF) is limited by multiple adverse effects, including renal toxicity leading to acute kidney injury. In mice with DCLF-induced nephrotoxicity, TDZD-8, a selective glycogen synthase kinase (GSK)3β inhibitor, improved acute kidney dysfunction and ameliorated tubular necrosis and apoptosis associated with induced cortical expression of cyclooxygenase-2 (COX-2) and prostaglandin E2. This renoprotective effect was blunted but still largely preserved in COX-2-null mice, suggesting that other GSK3β targets beyond COX-2 functioned in renal protection. Indeed, TDZD-8 diminished the mitochondrial permeability transition in DCLF-injured kidneys. In vitro, GSK3β inhibition reinstated viability and suppressed necrosis and apoptosis in DCLF-stimulated tubular epithelial cells. DCLF elicited oxidative stress, enhanced the activity of the redox-sensitive GSK3β, and promoted a mitochondrial permeability transition by interacting with cyclophilin D, a key component of the mitochondrial permeability transition pore. TDZD-8 blocked GSK3β activity and prevented GSK3β-mediated cyclophilin D phosphorylation and the ensuing mitochondrial permeability transition, concomitant with normalization of intracellular ATP. Conversely, ectopic expression of a constitutively active GSK3β abolished the effects of TDZD-8. Hence, inhibition of GSK3β ameliorates NSAID-induced acute kidney injury by induction of renal cortical COX-2 and direct inhibition of the mitochondrial permeability transition.
Collapse
Affiliation(s)
- Hao Bao
- Research Institute of Nephrology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Yan Ge
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Shougang Zhuang
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Lance D Dworkin
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Zhihong Liu
- Research Institute of Nephrology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Brown University School of Medicine, Providence, Rhode Island, USA
| |
Collapse
|
14
|
McKenzie JA, Bixby EC, Silva MJ. Differential gene expression from microarray analysis distinguishes woven and lamellar bone formation in the rat ulna following mechanical loading. PLoS One 2011; 6:e29328. [PMID: 22216249 PMCID: PMC3245266 DOI: 10.1371/journal.pone.0029328] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Accepted: 11/26/2011] [Indexed: 12/21/2022] Open
Abstract
Formation of woven and lamellar bone in the adult skeleton can be induced through mechanical loading. Although much is known about the morphological appearance and structural properties of the newly formed bone, the molecular responses to loading are still not well understood. The objective of our study was to use a microarray to distinguish the molecular responses between woven and lamellar bone formation induced through mechanical loading. Rat forelimb loading was completed in a single bout to induce the formation of woven bone (WBF loading) or lamellar bone (LBF loading). A set of normal (non-loaded) rats were used as controls. Microarrays were performed at three timepoints after loading: 1 hr, 1 day and 3 days. Confirmation of microarray results was done for a select group of genes using quantitative real-time PCR (qRT-PCR). The micorarray identified numerous genes and pathways that were differentially regulated for woven, but not lamellar bone formation. Few changes in gene expression were evident comparing lamellar bone formation to normal controls. A total of 395 genes were differentially expressed between formation of woven and lamellar bone 1 hr after loading, while 5883 and 5974 genes were differentially expressed on days 1 and 3, respectively. Results suggest that not only are the levels of expression different for each type of bone formation, but that distinct pathways are activated only for woven bone formation. A strong early inflammatory response preceded an increase in angiogenic and osteogenic gene expression for woven bone formation. Furthermore, at later timepoints there was evidence of bone resorption after WBF loading. In summary, the vast coverage of the microarray offers a comprehensive characterization of the early differences in expression between woven and lamellar bone formation.
Collapse
Affiliation(s)
- Jennifer A McKenzie
- Department of Orthopaedics, Washington University, St. Louis, Missouri, USA.
| | | | | |
Collapse
|
15
|
Kesavan C, Wergedal JE, Lau KHW, Mohan S. Conditional disruption of IGF-I gene in type 1α collagen-expressing cells shows an essential role of IGF-I in skeletal anabolic response to loading. Am J Physiol Endocrinol Metab 2011; 301:E1191-7. [PMID: 21878662 PMCID: PMC3233773 DOI: 10.1152/ajpendo.00440.2011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To establish a causal role for locally produced IGF-I in the mechanical strain response in the bone, we have generated mice with conditional disruption of the insulin-like growth factor (IGF) I gene in type 1α(2) collagen-expressing cells using the Cre-loxP approach. At 10 wk of age, loads adjusted to account for bone size difference were applied via four-point bending or axial loading (AL) in mice. Two wk of bending and AL produced significant increases in bone mineral density and bone size at the middiaphysis of wild-type (WT), but not knockout (KO), mice. In addition, AL produced an 8-25% increase in trabecular parameters (bone volume-tissue volume ratio, trabecular thickness, and trabecular bone mineral density) at the secondary spongiosa of WT, but not KO, mice. Histomorphometric analysis at the trabecular site revealed that AL increased osteoid width by 60% and decreased tartrate-resistance acidic phosphatase-labeled surface by 50% in the WT, but not KO, mice. Consistent with the in vivo data, blockade of IGF-I action with inhibitory IGF-binding protein (IGFBP4) in vitro completely abolished the fluid flow stress-induced MC3T3-E1 cell proliferation. One-way ANOVA revealed that expression levels of EFNB1, EFNB2, EFNA2, EphB2, and NR4a3 were different in the loaded bones of WT vs. KO mice and may, in part, be responsible for the increase in bone response to loading in the WT mice. In conclusion, IGF-I expressed in type 1 collagen-producing bone cells is critical for converting mechanical signal to anabolic signal in bone, and other growth factors cannot compensate for the loss of local IGF-I.
Collapse
Affiliation(s)
- Chandrasekhar Kesavan
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California, USA
| | | | | | | |
Collapse
|
16
|
Kamel MA, Picconi JL, Lara-Castillo N, Johnson ML. Activation of β-catenin signaling in MLO-Y4 osteocytic cells versus 2T3 osteoblastic cells by fluid flow shear stress and PGE2: Implications for the study of mechanosensation in bone. Bone 2010; 47:872-81. [PMID: 20713195 PMCID: PMC2952691 DOI: 10.1016/j.bone.2010.08.007] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 07/19/2010] [Accepted: 08/10/2010] [Indexed: 12/20/2022]
Abstract
The osteocyte is hypothesized to be the mechanosensory cell in bone. However, osteoblastic cell models have been most commonly used to investigate mechanisms of mechanosensation in bone. Therefore, we sought to determine if differences might exist between osteocytic and osteoblastic cell models relative to the activation of β-catenin signaling in MLO-Y4 osteocytic, 2T3 osteoblastic and primary neonatal calvarial cells (NCCs) in response to pulsatile fluid flow shear stress (PFFSS). β-catenin nuclear translocation was observed in the MLO-Y4 cells at 2 and 16 dynes/cm(2) PFFSS, but only at 16 dynes/cm(2) in the 2T3 or NCC cultures. The MLO-Y4 cells released high amounts of PGE(2) into the media at all levels of PFFSS (2-24 dynes/cm(2)) and we observed a biphasic pattern relative to the level of PFFSS. In contrast PGE(2) release by 2T3 cells was only detected during 16 and 24 dynes/cm(2) PFFSS starting at >1h and never reached the levels produced by the MLO-Y4 cells. Exogenously added PGE(2) was able to induce β-catenin nuclear translocation in all cells suggesting that the differences between the cell lines observed for β-catenin nuclear translocation were associated with the differences in PGE(2) production. To investigate a possible mechanism for the differences in PGE(2) release by the MLO-Y4 and 2T3 cells we examined the regulation of Ptgs2 (Cox-2) gene expression by PFFSS. 2T3 cell Ptgs2 mRNA levels at both 0 and 24h after 2h of PFFSS showed biphasic increases with peaks at 4 and 24 dynes/cm(2) and 24-hour levels were higher than zero-hour levels. MLO-Y4 cell Ptgs2 expression was similarly biphasic; however at 24-hour post-flow Ptgs2 mRNA levels were lower. Our data suggest significant differences in the sensitivity and kinetics of the response mechanisms of the 2T3 and neonatal calvarial osteoblastic versus MLO-Y4 osteocytic cells to PFFSS. Furthermore our data support a role for PGE(2) in mediating the activation of β-catenin signaling in response to the fluid flow shear stress.
Collapse
Affiliation(s)
- Mohamed A. Kamel
- Department of Oral Biology, UMKC School of Dentistry, 650 East 25 Street, Kansas City, MO 64108
| | - Jason L. Picconi
- Department of Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242
| | - Nuria Lara-Castillo
- Department of Oral Biology, UMKC School of Dentistry, 650 East 25 Street, Kansas City, MO 64108
| | - Mark L. Johnson
- Department of Oral Biology, UMKC School of Dentistry, 650 East 25 Street, Kansas City, MO 64108
| |
Collapse
|
17
|
Saunders MM, Simmerman LA, Reed GL, Sharkey NA, Taylor AF. Biomimetic bone mechanotransduction modeling in neonatal rat femur organ cultures: structural verification of proof of concept. Biomech Model Mechanobiol 2010; 9:539-50. [PMID: 20169394 PMCID: PMC2908729 DOI: 10.1007/s10237-010-0195-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 01/22/2010] [Indexed: 10/19/2022]
Abstract
The goal of this work was to develop and validate a whole bone organ culture model to be utilized in biomimetic mechanotransduction research. Femurs harvested from 2-day-old neonatal rat pups were maintained in culture for 1 week post-harvest and assessed for growth and viability. For stimulation studies, femurs were physiologically stimulated for 350 cycles 24 h post-harvest then maintained in culture for 1 week at which time structural tests were conducted. Comparing 1 and 8 days in culture, bones grew significantly in size over the 7-day culture period. In addition, histology supported adequate diffusion and organ viability at 2 weeks in culture. For stimulation studies, 350 cycles of physiologic loading 24 h post-harvest resulted in increased bone strength over the 7-day culture period. In this work, structural proof of concept was established for the use of whole bone organ cultures as mechanotransduction models. Specifically, this work established that these cultures grow and remain viable in culture, are adequately nourished via diffusion and are capable of responding to a brief bout of mechanical stimulation with an increase in strength.
Collapse
|
18
|
Xu M, Choudhary S, Voznesensky O, Gao Q, Adams D, Diaz-Doran V, Wu Q, Goltzman D, Raisz LG, Pilbeam CC. Basal bone phenotype and increased anabolic responses to intermittent parathyroid hormone in healthy male COX-2 knockout mice. Bone 2010; 47:341-52. [PMID: 20471507 PMCID: PMC3056501 DOI: 10.1016/j.bone.2010.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 04/26/2010] [Accepted: 05/06/2010] [Indexed: 01/09/2023]
Abstract
Cyclooxygenase-2 (COX-2) knockout (KO) mice in inbred strains can have renal dysfunction with secondary hyperparathyroidism (HPTH), making direct effects of COX-2 KO on bone difficult to assess. COX-2 KO mice in an outbred CD-1 background did not have renal dysfunction but still had two-fold elevated PTH compared to wild type (WT) mice. Compared to WT mice, KO mice had increased serum markers of bone turnover, decreased femoral bone mineral density (BMD) and cortical bone thickness, but no differences in trabecular bone volume by microCT or dynamic histomorphometry. Because PTH is a potent inducer of COX-2 and prostaglandin (PG) production, we examined the effects of COX-2 KO on bone responses after 3 weeks of intermittent PTH. Intermittent PTH increased femoral BMD and cortical bone area more in KO mice than in WT mice and increased trabecular bone volume in the distal femur in both WT and KO mice. Although not statistically significant, PTH-stimulated increases in trabecular bone tended to be greater in KO mice than in WT mice. PTH increased serum markers of bone formation and resorption more in KO than in WT mice but increased the ratio of osteoblastic surface-to-osteoclastic surface only in KO mice. PTH also increased femoral mineral apposition rates and bone formation rates in KO mice more than in WT mice. Acute mRNA responses to PTH of genes that might mediate some anabolic and catabolic effects of PTH tended to be greater in KO than WT mice. We conclude that (1) the basal bone phenotype in male COX-2 KO mice might reflect HPTH, COX-2 deficiency or both, and (2) increased responses to intermittent PTH in COX-2 KO mice, despite the presence of chronic HPTH, suggest that absence of COX-2 increased sensitivity to PTH. It is possible that manipulation of endogenous PGs could have important clinical implications for anabolic therapy with PTH.
Collapse
Affiliation(s)
- Manshan Xu
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kohrt WM, Barry DW, Van Pelt RE, Jankowski CM, Wolfe P, Schwartz RS. Timing of ibuprofen use and bone mineral density adaptations to exercise training. J Bone Miner Res 2010; 25:1415-22. [PMID: 20200939 PMCID: PMC3152206 DOI: 10.1002/jbmr.24] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Prostaglandins (PGs) are essential signaling factors in bone mechanotransduction. In animals, inhibition of the enzyme responsible for PG synthesis (cyclooxygenase) by nonsteroidal anti-inflammatory drugs (NSAIDs) blocks the bone-formation response to loading when administered before, but not immediately after, loading. The aim of this proof-of-concept study was to determine whether the timing of NSAID use influences bone mineral density (BMD) adaptations to exercise in humans. Healthy premenopausal women (n = 73) aged 21 to 40 years completed a supervised 9-month weight-bearing exercise training program. They were randomized to take (1) ibuprofen (400 mg) before exercise, placebo after (IBUP/PLAC), (2) placebo before, ibuprofen after (PLAC/IBUP), or (3) placebo before and after (PLAC/PLAC) exercise. Relative changes in hip and lumbar spine BMD from before to after exercise training were assessed using a Hologic Delphi-W dual-energy X-ray absorptiometry (DXA) instrument. Because this was the first study to evaluate whether ibuprofen use affects skeletal adaptations to exercise, only women who were compliant with exercise were included in the primary analyses (IBUP/PLAC, n = 17; PLAC/PLAC, n = 23; and PLAC/IBUP, n = 14). There was a significant effect of drug treatment, adjusted for baseline BMD, on the BMD response to exercise for regions of the hip (total, p < .001; neck, p = .026; trochanter, p = .040; shaft, p = .019) but not the spine (p = .242). The largest increases in BMD occurred in the group that took ibuprofen after exercise. Total-hip BMD changes averaged -0.2% +/- 1.3%, 0.4% +/- 1.8%, and 2.1% +/- 1.7% in the IBUP/PLAC, PLAC/PLAC, and PLAC/IBUP groups, respectively. This preliminary study suggests that taking NSAIDs after exercise enhances the adaptive response of BMD to exercise, whereas taking NSAIDs before may impair the adaptive response.
Collapse
Affiliation(s)
- Wendy M Kohrt
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Denver, Denver, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Affiliation(s)
- Chenyu Huang
- Department of Plastic, Reconstructive and Aesthetic SurgeryNippon Medical School Tokyo Japan
- Department of Plastic SurgeryMeitan General Hospital Beijing China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic SurgeryNippon Medical School Tokyo Japan
| |
Collapse
|
21
|
Blackwell KA, Raisz LG, Pilbeam CC. Prostaglandins in bone: bad cop, good cop? Trends Endocrinol Metab 2010; 21:294-301. [PMID: 20079660 PMCID: PMC2862787 DOI: 10.1016/j.tem.2009.12.004] [Citation(s) in RCA: 200] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 12/10/2009] [Accepted: 12/11/2009] [Indexed: 02/01/2023]
Abstract
Prostaglandins (PGs) are multifunctional regulators of bone metabolism that stimulate both bone resorption and formation. PGs have been implicated in bone resorption associated with inflammation and metastatic bone disease, and also in bone formation associated with fracture healing and heterotopic ossification. Recent studies have identified roles for inducible cyclooxygenase (COX)-2 and PGE(2) receptors in these processes. Although the effects of PGs have been most often associated with cAMP production and protein kinase A activation, PGs can engage an extensive G-protein signaling network. Further analysis of COX-2 and PG receptors and their downstream G-protein signaling in bone could provide important clues to the regulation of skeletal cell growth in both health and disease.
Collapse
Affiliation(s)
- Katherine A Blackwell
- New England Musculoskeletal Institute, University of Connecticut Health Center, 263 Farmington Ave, Farmington, CT, USA
| | | | | |
Collapse
|
22
|
Chen JC, Beaupré GS, Carter DR. An approach to quantifying bone overloading and hypertrophy with applications to multiple experimental studies. Bone 2010; 46:322-9. [PMID: 19800044 DOI: 10.1016/j.bone.2009.09.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Revised: 08/23/2009] [Accepted: 09/23/2009] [Indexed: 10/20/2022]
Abstract
Many studies have investigated mechanically induced bone formation in mice and rats by applying loads to the long bones, and measuring changes in periosteal cortical bone apposition rates. However, the results are difficult to compare among each other because the loading schemes are generally different. The purpose of the present study was to develop a theoretical framework for evaluating the mechanical stimulus based on the bone daily strain stimulus, which is a function of loading cycles and bone strains. The daily strain stimulus would act as a single unifying parameter for directly comparing data from existing in vivo experiments, and is applied here to twenty previous rat and mouse studies. To calculate the daily strain stimulus, we determined the periosteal daily strain stimulus necessary for bone maintenance (xi(peri,0)) and the strain-cycle weighting exponent (m). In the first approach, we applied data from Rubin and Lanyon's bone maintenance studies. We calculated xi(peri,0) to be 2793 microstrain/day, and m to be 4.5. In the second approach, we used Fritton et al. 's strain gage recordings to calculate xi(peri,0) to be 1496 microstrain/day, and used an m value of 11.88, equal to human bone compressive fatigue properties. Fatigue data provided physiological relevance, and was useful for applying an established remodeling theory to in vivo studies. For both approaches, xi(peri,0) was below the fracture level. We then analyzed the applied strains, cycles, and periosteal bone apposition rates from the previous studies. The range of daily strain stimuli calculated using the first approach was much larger than the range using the second approach (2793-17312 microstrain/day compared to 1496-7681 microstrain/day). None of the studies applied a daily strain stimulus above the complete fatigue failure level, but some studies applied loading that could result in major fatigue microdamage. Bone apposition rates generally increased with increasing daily strain stimulus, which was consistent with previous theoretical models. The results suggest that the daily strain stimulus may be a reasonable first approximation for predicting bone apposition rates in a consistent manner. The use of the daily strain stimulus may be helpful for improving the design of future bone loading studies.
Collapse
Affiliation(s)
- J C Chen
- Bone & Joint Rehabilitation R&D Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.
| | | | | |
Collapse
|
23
|
Papachristou DJ, Papachroni KK, Basdra EK, Papavassiliou AG. Signaling networks and transcription factors regulating mechanotransduction in bone. Bioessays 2009; 31:794-804. [PMID: 19444851 DOI: 10.1002/bies.200800223] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mechanical stimulation has a critical role in the development and maintenance of the skeleton. This function requires the perception of extracellular stimuli as well as their conversion into intracellular biochemical responses. This process is called mechanotransduction and is mediated by a plethora of molecular events that regulate bone metabolism. Indeed, mechanoreceptors, such as integrins, G protein-coupled receptors, receptor protein tyrosine kinases, and stretch-activated Ca(2+) channels, together with their downstream effectors coordinate the transmission of load-induced signals to the nucleus and the expression of bone-related genes. During the past decade, scientists have gained increasing insight into the molecular networks implicated in bone mechanotransduction. In the present paper, we consider the major signaling cascades and transcription factors that control bone and cartilage mechanobiology and discuss the influence of the mechanical microenvironment on the determination of skeletal morphology.
Collapse
|
24
|
Wallace JM, Rajachar RM, Allen MR, Bloomfield SA, Robey PG, Young MF, Kohn DH. Exercise-induced changes in the cortical bone of growing mice are bone- and gender-specific. Bone 2007; 40:1120-7. [PMID: 17240210 PMCID: PMC2729655 DOI: 10.1016/j.bone.2006.12.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 11/02/2006] [Accepted: 12/01/2006] [Indexed: 10/23/2022]
Abstract
Fracture risk and mechanical competence of bone are functions of bone mass and tissue quality, which in turn are dependent on the bone's mechanical environment. Male mice have a greater response to non-weight-bearing exercise than females, resulting in larger, stronger bones compared with control animals. The aim of this study was to test the hypothesis that short-term weight-bearing running during growth (21 days starting at 8 weeks of age; 30 min/day; 12 m/min; 5 degrees incline; 7 days/week) would similarly have a greater impact on cross-sectional geometry and mechanical competence in the femora and tibiae of male mice versus females. Based on the orientation of the legs during running and the proximity of the tibia to the point of impact, this response was hypothesized to be greatest in the tibia. Exercise-related changes relative to controls were assayed by four-point bending tests, while volumetric bone mineral density and cross-sectional geometry were also assessed. The response to running was bone- and gender-specific, with male tibiae demonstrating the greatest effects. In male tibiae, periosteal perimeter, endocortical perimeter, cortical area, medial-lateral width and bending moment of inertia increased versus control mice suggesting that while growth is occurring in these mice between 8 and 11 weeks of age, exercise accelerated this growth resulting in a greater increase in bone tissue over the 3 weeks of the study. Exercise increased tissue-level strain-to-failure and structural post-yield deformation in the male tibiae, but these post-yield benefits came at the expense of decreased yield deformation, structural and tissue-level yield strength and tissue-level ultimate strength. These results suggest that exercise superimposed upon growth accelerated growth-related increases in tibial cross-sectional dimensions. Exercise also influenced the quality of this forming bone, significantly impacting structural and tissue-level mechanical properties.
Collapse
Affiliation(s)
- Joseph M. Wallace
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Rupak M. Rajachar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Matthew R. Allen
- Texas A&M University, Department of Health and Kinesiology; College Station, TX
| | - Susan A. Bloomfield
- Texas A&M University, Department of Health and Kinesiology; College Station, TX
| | - Pamela G. Robey
- National Institutes of Health, National Institute of Dental and Craniofacial Research, Craniofacial and Skeletal Diseases Branch, Department of Health and Human Services; Bethesda, MD
| | - Marian F. Young
- National Institutes of Health, National Institute of Dental and Craniofacial Research, Craniofacial and Skeletal Diseases Branch, Department of Health and Human Services; Bethesda, MD
| | - David H. Kohn
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI
- National Institutes of Health, National Institute of Dental and Craniofacial Research, Craniofacial and Skeletal Diseases Branch, Department of Health and Human Services; Bethesda, MD
- Corresponding Author: David H. Kohn, Ph.D., University of Michigan, Department of Biologic & Materials Sciences, 1011 N. University Ave., Ann Arbor, MI 48109-1078, Ph: (734) 764-2206, Fax: (734) 647-2110, E-mail:
| |
Collapse
|
25
|
Zhang P, Tanaka SM, Sun Q, Turner CH, Yokota H. Frequency-dependent enhancement of bone formation in murine tibiae and femora with knee loading. J Bone Miner Metab 2007; 25:383-91. [PMID: 17968490 PMCID: PMC2902372 DOI: 10.1007/s00774-007-0774-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Accepted: 05/10/2007] [Indexed: 12/01/2022]
Abstract
Knee loading is a relatively new loading modality in which dynamic loads are laterally applied to the knee to induce bone formation in the tibia and the femur. The specific aim of the current study was to evaluate the effects of loading frequencies (in Hz) on bone formation at the site away from the loading site on the knee. The left knee of C57/BL/6 mice was loaded with 0.5 N force at 5, 10, or 15 Hz for 3 min/day for 3 consecutive days, and bone histomorphometry was conducted at the site 75% away from the loading site along the length of tibiae and femora. The results revealed frequency-dependent induction of bone formation, in which the dependence was different in the tibia and the femur. Compared with the sham-loading control, for instance, the cross-sectional cortical area was elevated maximally at 5 Hz in the tibia, whereas the most significant increase was observed at 15 Hz in the femur. Furthermore, mineralizing surface, mineral apposition rate, and bone formation rate were the highest at 5 Hz in the tibia (2.0-, 1.4-, and 2.7 fold, respectively) and 15 Hz in the femur (1.5-, 1.2-, and 1.8 fold, respectively). We observed that the tibia had a lower bone mineral density with more porous microstructures than the femur. Those differences may contribute to the observed differential dependence on loading frequencies.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Biomedical Engineering, Indiana University - Purdue University Indianapolis (IUPUI), Fesler Hall 115, 1120 South Drive, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
26
|
Robertson G, Xie C, Chen D, Awad H, Schwarz EM, O’Keefe RJ, Guldberg RE, Zhang X. Alteration of femoral bone morphology and density in COX-2-/- mice. Bone 2006; 39:767-72. [PMID: 16731065 PMCID: PMC2647994 DOI: 10.1016/j.bone.2006.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Revised: 02/20/2006] [Accepted: 04/04/2006] [Indexed: 02/03/2023]
Abstract
A role of COX-2 in pathological bone destruction and fracture repair has been established; however, few studies have been conducted to examine the involvement of COX-2 in maintaining bone mineral density and bone micro-architecture. In this study, we examined bone morphology in multiple trabecular and cortical regions within the distal and diaphyseal femur of 4-month-old wild-type and COX-2-/- mice using micro-computed tomography. Our results demonstrated that while COX-2-/- female mice had normal bone geometry and trabecular microarchitecture at 4 months of age, the male knockout mice displayed reduced bone volume fraction within the distal femoral metaphysis. Furthermore, male COX-2-/- mice had a significant reduction in cortical bone mineral density within the central cortical diaphysis and distal epiphysis and metaphysis. Consistent with the observed reduction in cortical mineral density, biomechanical testing via 4-point-bending showed that male COX-2-/- mice had a significant increase in postyield deformation, indicating a ductile bone phenotype in male COX-2-/- mice. In conclusion, our study suggests that genetic ablation of COX-2 may have a sex-related effect on cortical bone homeostasis and COX-2 plays a role in maintaining normal bone micro-architecture and density in mice.
Collapse
Affiliation(s)
- Galen Robertson
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Di Chen
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Hani Awad
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Regis J. O’Keefe
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| |
Collapse
|
27
|
Liedert A, Kaspar D, Blakytny R, Claes L, Ignatius A. Signal transduction pathways involved in mechanotransduction in bone cells. Biochem Biophys Res Commun 2006; 349:1-5. [PMID: 16930556 DOI: 10.1016/j.bbrc.2006.07.214] [Citation(s) in RCA: 201] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 07/24/2006] [Indexed: 11/17/2022]
Abstract
Several in vivo and in vitro studies with different loading regimens showed that mechanical stimuli have an influence on proliferation and differentiation of bone cells. Prerequisite for this influence is the transduction of mechanical signals into the cell, a phenomenon that is termed mechanotransduction, which is essential for the maintenance of skeletal homeostasis in adults. Mechanoreceptors, such as the integrins, cadherins, and stretch-activated Ca2+ channels, together with various signal transduction pathways, are involved in the mechanotransduction process that ultimately regulates gene expression in the nucleus. Mechanotransduction itself is considered to be regulated by hormones, the extracellular matrix of the osteoblastic cells and the mode of the mechanical stimulus.
Collapse
Affiliation(s)
- Astrid Liedert
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Germany.
| | | | | | | | | |
Collapse
|
28
|
Grimston SK, Screen J, Haskell JH, Chung DJ, Brodt MD, Silva MJ, Civitelli R. Role of connexin43 in osteoblast response to physical load. Ann N Y Acad Sci 2006; 1068:214-24. [PMID: 16831921 DOI: 10.1196/annals.1346.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Gap junctions are hexameric transmembrane channels formed by connexins, and are responsible for direct cell-to-cell communication. The most abundant gap junction protein in bone is connexin43 (Cx43), although connexin45 (Cx45) is also expressed. In the present study, we tested the hypothesis that bone cell responses to mechanical stimulation are dependent on the type of gap junction communication provided by Cx43 in vitro and in an in vivo model of physical load. Application of cyclic stretch to calvaria osteoblasts results in a modest but detectable increase in PGE2 levels, and the amount of PGE2 produced was lower in cells isolated from Cx43 null mice. Mice with an osteoblast-specific deletion of the Cx43 gene were subjected to an in vivo four-point bending protocol on the tibia. This resulted in fast and exuberant formation of woven bone at the region directly below the loading fulcrum in both osteoblast Cx43-deleted and wild-type mice. However, indirect measurement of endosteal bone apposition suggested a less pronounced effect of physical load in Cx43-deficient than in wild-type mice. Taken together, these results indicate that deficiency of Cx43 in osteoblasts attenuates but does not abolish anabolic responses to mechanical strain.
Collapse
Affiliation(s)
- Susan K Grimston
- Division of Bone and Mineral Diseases, Washington University in St. Louis, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Liedert A, Kaspar D, Claes L, Ignatius A. Signal transduction pathways involved in mechanical regulation of HB-GAM expression in osteoblastic cells. Biochem Biophys Res Commun 2006; 342:1070-6. [PMID: 16513091 DOI: 10.1016/j.bbrc.2006.02.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Accepted: 02/13/2006] [Indexed: 11/25/2022]
Abstract
Protein kinase C (PKC), protein kinase A (PKA), prostaglandin synthesis, and various mitogen-activated protein kinases (MAPKs) have been reported to be activated in bone cells by mechanical loading. We studied the involvement of these signal transduction pathways in the downregulation of HB-GAM expression in osteoblastic cells after cyclic stretching. Specific antagonists and agonists of these signal transduction pathways were added to cells before loading and to non-loaded control cells. Quantitative RT-PCR was used to evaluate gene expression. The data demonstrated that the extracellular signal-regulated kinase (ERK) 1/2 pathway, PKC, PKA, p38, and c-Jun N-terminal kinase MAPK participated in the mechanical downregulation of HB-GAM expression, whereas prostaglandin synthesis did not seem to be involved.
Collapse
Affiliation(s)
- Astrid Liedert
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Germany.
| | | | | | | |
Collapse
|
30
|
Mehrotra M, Saegusa M, Voznesensky O, Pilbeam C. Role of Cbfa1/Runx2 in the fluid shear stress induction of COX-2 in osteoblasts. Biochem Biophys Res Commun 2006; 341:1225-30. [PMID: 16476583 DOI: 10.1016/j.bbrc.2006.01.084] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 01/20/2006] [Indexed: 10/25/2022]
Abstract
Induction of cyclooxygenase-2 (COX-2) is thought to be important for the anabolic effects of mechanical loading. The transcription factor Cbfa1/Runx2 is essential for osteoblastic differentiation. We examined the role of Cbfa1 in the fluid shear stress (FSS) induction of COX-2 in MC3T3-E1 cells stably transfected with a COX-2 promoter-luciferase reporter. Cells were subjected to FSS for 30 min and returned to static culture (post-FSS). COX-2 mRNA and promoter activity peaked 0.5-1h and 2-3h, respectively, post-FSS. Mutation of the Cbfa1 consensus sequence at -267/-261 bp decreased the FSS fold-induction of luciferase activity by 50%. On electrophoretic mobility shift assay (EMSA), proteins binding to an oligonucleotide spanning the Cbfa1 site were supershifted by specific antibody to Cbfa1. FSS did not increase Cbfa1 binding on EMSA or Cbfa1 mRNA or protein levels. These data suggest that transcriptional activity of Cbfa1, independent of its level of expression, is necessary for maximal FSS induction of COX-2 in osteoblasts.
Collapse
Affiliation(s)
- Meenal Mehrotra
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | |
Collapse
|
31
|
Abstract
The major goal of clinicians when treating acute musculoskeletal injuries is to return athletes to their pre-injury level of function, ideally in the shortest time possible and without compromising tissue-level healing. In this regard, a commonly used intervention is the taking of NSAIDs. These are used to limit the amount and duration of inflammation, and to control pain. While NSAIDs have become synonymous with the management of acute musculoskeletal injuries, their efficacy has yet to be proven. This is of particular concern in view of recent research investigating the latest class of NSAIDs - selective cyclo-oxygenase-2 inhibitors (COXIBs). COXIBs were developed to reduce the adverse gastrointestinal (GI) effects of traditional NSAIDs. While they have beneficial anti-inflammatory and analgesic properties, and appear to facilitate earlier return to function following acute injury, the effect of COXIBs on tissue-level healing is currently unknown. In experimental animal models of acute injury, COXIBs have been shown to be detrimental to tissue-level repair. Specifically, they have been shown to impair mechanical strength return following acute injury to bone, ligament and tendon. Clinically, this may have implications for ongoing morbidity and future injury susceptibility. However, the current animal studies have limited translation to the clinical setting, particularly because of significant limitations relating to drug use and dosage in these studies. There is currently no randomised, controlled trial evidence of the tissue-level effects of COXIBs on acute musculoskeletal injuries. In addition to questions relating to the effect of COXIBs on tissue-level healing, further questions regarding the use of these agents have been raised given a recent link being shown between one COXIB (rofecoxib) and an increased risk for adverse cardiovascular events. Whether this finding is related to the individual properties of rofecoxib or is a class phenomenon is the subject of ongoing investigation. However, in light of the potential risks associated with using COXIBs, an acceptable and possibly safer alternative in the management of acute musculoskeletal injuries may be to use traditional NSAIDs. Traditional NSAIDs do carry the potential for greater adverse GI effects and their clinical effects on tissue-level healing remain relatively unknown. However, they do not appear to be associated with adverse cardiovascular effects, and they are effective pain relievers and cheaper alternatives.
Collapse
Affiliation(s)
- Stuart J Warden
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, 46202, USA.
| |
Collapse
|