1
|
Tauer JT, Thiele T, Julien C, Ofer L, Zaslansky P, Shahar R, Willie BM. Swim training induces distinct osseous gene expression patterns in anosteocytic and osteocytic teleost fish. Bone 2024; 185:117125. [PMID: 38754573 DOI: 10.1016/j.bone.2024.117125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
The traditional understanding of bone mechanosensation implicates osteocytes, canaliculi, and the lacunocanalicular network in biomechanical adaptation. However, recent findings challenge this notion, as shown in advanced teleost fish where anosteocytic bone lacking osteocytes are nevertheless responsive to mechanical load. To investigate specific molecular mechanisms involved in bone mechanoadaptation in osteocytic and anosteocytic fish bone, we conducted a 5-min single swim-training experiment with zebrafish and ricefish, respectively. Through RNASeq analysis of fish spines, analyzed at various time points following swim training, we uncovered distinct gene expression patterns in osteocytic and anosteocytic fish bones. Notably, osteocytic fish bone exhibited an early response to mechanical load, contrasting to a delayed response observed in anosteocytic fish bones, both within 8 h following stimulation. We identified an increase in osteoblast differentiation in anosteocytic bone following training, while chordoblast activity was delayed. This temporal response suggests a time-dependent adaptation in anosteocytic bone, indicating the presence of intricate feedback networks within bone that lacks osteocytes.
Collapse
Affiliation(s)
- Josephine T Tauer
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada; Department of Pediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Tobias Thiele
- Julius Wolff Institute and Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Catherine Julien
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Lior Ofer
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University, Rehovot, Israel
| | - Paul Zaslansky
- Department of Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ron Shahar
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University, Rehovot, Israel
| | - Bettina M Willie
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada.
| |
Collapse
|
2
|
Xu C, Ji G, Chen X, Yan L, Liang T, Liu J, Wang F. Sclerostin antibody promotes bone formation through the Wnt/β-catenin signaling pathway in femoral trochlear after patellar instability. Connect Tissue Res 2023; 64:148-160. [PMID: 36379907 DOI: 10.1080/03008207.2022.2135507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE The molecular mechanism of patellar instability (PI) remains unknown. The purpose of this study was to explore the function of SOST/sclerostin in PI and examine the effect of sclerostin antibody (Scl-Ab). MATERIALS AND METHODS We randomly divided 60 male 3-week-old C57Bl/6 mice into four groups: sham, PI, Scl-Ab intraperitoneal injection (Scl-Ab IP), Scl-Ab intraarticular injection (Scl-Ab IA). PI was established in the latter three groups. The Scl-Ab IP/IA groups were administered with an intraperitoneal/intraarticular Scl-Ab injection (100 mg/kg, 20 µl), respectively, at 5-day intervals. Distal femurs were collected 30 days after the surgery. The SOST/sclerostin, β-catenin, ALP, OPG and RANKL expression in distal femur were determined. Trochlear morphology and structural parameters of the trabecular and cortical bone compartments were determined by micro-CT. Further sub-regional analysis was performed. HE staining and Masson's trichrome staining were performed to evaluate cartilage changes. RESULTS PI increased the expression of SOST/sclerostin and RANKL, and decreased β-catenin, ALP and OPG levels, while Scl-Ab IP reversed these changes. Scl-Ab IP brought trochlear morphology closer to normality. Additionally, Scl-Ab IP significantly improved most of the bone parameters. Importantly, both PI and Scl-Ab IP acted mainly on trabecular bone. Histological analysis showed that Scl-Ab IP protected cartilage from degeneration. However, Scl-Ab IA did not protect against bone loss or cartilage degradation. CONCLUSIONS SOST/sclerostin plays an important role in PI and systemic Scl-Ab use promotes bone formation through the Wnt/β-catenin signaling pathway in the femoral trochlear after PI.
Collapse
Affiliation(s)
- Chenyue Xu
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| | - Gang Ji
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| | - Xiaobo Chen
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| | - Lirong Yan
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tuwan Liang
- College of Medical, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Junle Liu
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fei Wang
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
3
|
Chlebek C, Rosen CJ. The Role of Bone Cell Energetics in Altering Bone Quality and Strength in Health and Disease. Curr Osteoporos Rep 2023; 21:1-10. [PMID: 36435911 DOI: 10.1007/s11914-022-00763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Bone quality and strength are diminished with age and disease but can be improved by clinical intervention. Energetic pathways are essential for cellular function and drive osteogenic signaling within bone cells. Altered bone quality is associated with changes in the energetic activity of bone cells following diet-based or therapeutic interventions. Energetic pathways may directly or indirectly contribute to changes in bone quality. The goal of this review is to highlight tissue-level and bioenergetic changes in bone health and disease. RECENT FINDINGS Bone cell energetics are an expanding field of research. Early literature primarily focused on defining energetic activation throughout the lifespan of bone cells. Recent studies have begun to connect bone energetic activity to health and disease. In this review, we highlight bone cell energetic demands, the effect of substrate availability on bone quality, altered bioenergetics associated with disease treatment and development, and additional biological factors influencing bone cell energetics. Bone cells use several energetic pathways during differentiation and maturity. The orchestration of bioenergetic pathways is critical for healthy cell function. Systemic changes in substrate availability alter bone quality, potentially due to the direct effects of altered bone cell bioenergetic activity. Bone cell bioenergetics may also contribute directly to the development and treatment of skeletal diseases. Understanding the role of energetic pathways in the cellular response to disease will improve patient treatment.
Collapse
Affiliation(s)
- Carolyn Chlebek
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, USA.
| |
Collapse
|
4
|
Xu X, Yang H, Bullock WA, Gallant MA, Ohlsson C, Bellido TM, Main RP. Osteocyte Estrogen Receptor β (Ot-ERβ) Regulates Bone Turnover and Skeletal Adaptive Response to Mechanical Loading Differently in Male and Female Growing and Adult Mice. J Bone Miner Res 2023; 38:186-197. [PMID: 36321245 PMCID: PMC10108310 DOI: 10.1002/jbmr.4731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Age-related bone loss is a failure of balanced bone turnover and diminished skeletal mechanoadaptation. Estrogen receptors, ERα and ERβ, play critical roles in osteoprotective regulation activated by estrogen and mechanical signals. Previous studies mainly focused on ERα and showed that osteocyte-ERα (Ot-ERα) regulated trabecular, but not cortical bone, and played a minor role in load-induced cortical adaptation. However, the role of Ot-ERβ in bone mass regulation remains unrevealed. To address this issue, we characterized bone (re)modeling and gene expression in male and female mice with Ot-ERβ deletion (ERβ-dOT) and littermate control (LC) at 10 weeks (young) or 28 weeks (adult) of age, as well as their responses to in vivo tibial compressive loading. Increased cancellous bone mass appeared in the L4 vertebral body of young male ERβ-dOT mice. At the same time, femoral cortical bone gene expression showed signs consistent with elevated osteoblast and osteoclast activities (type-I collagen, Cat K, RANKL). Upregulated androgen receptor (AR) expression was observed in young male ERβ-dOT mice relative to LC, suggesting a compensatory effect of testosterone on male bone protection. In contrast, bone mass in L4 decreased in adult male ERβ-dOT mice, attributed to potentially increased bone resorption activity (Cat K) with no change in bone formation. There was no effect of ERβ-dOT on bone mass or gene expression in female mice. Sex-dependent regulation of Ot-ERβ also appeared in load-induced cortical responsiveness. Young female ERβ-dOT mice showed an enhanced tibial cortical anabolic adaptation compared with LC. In contrast, an attenuated cortical anabolic response presented at the proximal tibia in male ERβ-dOT mice at both ages. For the first time, our findings suggest that Ot-ERβ regulates bone (re)modeling and the response to mechanical signals through different mechanisms in males and females. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Xiaoyu Xu
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteINUSA
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| | - Haisheng Yang
- Department of Biomedical Engineering, Faculty of Environment and LifeBeijing University of TechnologyBeijingChina
| | | | - Maxim A. Gallant
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical NutritionInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Drug TreatmentSahlgrenska University HospitalGothenburgSweden
| | - Teresita M. Bellido
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockARUSA
| | - Russell P. Main
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteINUSA
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical SciencesPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
5
|
Zhang W, Xia CL, Ma JN, Li JX, Chen Q, Ou SJ, Yang Y, Qi Y, Xu CP. Effects of mitochondrial dysfunction on bone metabolism and related diseases: a scientometric study from 2003 to 2022. BMC Musculoskelet Disord 2022; 23:1016. [DOI: 10.1186/s12891-022-05911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/25/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
In recent years, mitochondrial dysfunction has been extensively studied and published, but research on the effects of mitochondrial dysfunction on bone metabolism and related diseases is only just beginning. Furthermore, no studies have been carried out to systematically illustrate this area from a scientometric point of view. The goal of this research is to review existing knowledge and identify new trends and possible hotspots in this area.
Methods
All publications related to the relationship between mitochondrial dysfunction and bone metabolism and related diseases from 2003 to 2022 were searched at the Web of Science Core Collection (WoSCC) on May 7, 2022. Four different analytical tools: VOSviewer 1.6.18, CiteSpace V 6.1, HistorCite (12.03.07), and Excel 2021 were used for the scientometric research.
Results
The final analysis included 555 valid records in total. Journal of Biological Chemistry (Co-citations = 916) is the most famous journal in this field. China (Percentage = 37%), the United States (Percentage = 24%), and Korea (Percentage = 12%) are the most productive countries. Blanco FJ and Choi EM are the main researchers with significant academic influence. Current research hotspots are basic research on mitochondrial dysfunction and the prevention or treatment of bone metabolism-related diseases.
Conclusion
The study of the consequences of mitochondrial dysfunction on bone metabolism and associated diseases is advancing rapidly. Several prominent researchers have published extensive literature and are widely cited. Future research in this area will focus on oxidative stress, aging, gene expression, and the pathogenesis of bone metabolism-related diseases.
Collapse
|
6
|
Chlebek C, Moore JA, Ross FP, van der Meulen MCH. Molecular Identification of Spatially Distinct Anabolic Responses to Mechanical Loading in Murine Cortical Bone. J Bone Miner Res 2022; 37:2277-2287. [PMID: 36054133 DOI: 10.1002/jbmr.4686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/05/2022] [Accepted: 08/20/2022] [Indexed: 11/08/2022]
Abstract
Osteoporosis affects over 200 million women worldwide, one-third of whom are predicted to suffer from an osteoporotic fracture in their lifetime. The most promising anabolic drugs involve administration of expensive antibodies. Because mechanical loading stimulates bone formation, our current data, using a mouse model, replicates the anabolic effects of loading in humans and may identify novel pathways amenable to oral treatment. Murine tibial compression produces axially varying deformations along the cortical bone, inducing highest strains at the mid-diaphysis and lowest at the metaphyseal shell. To test the hypothesis that load-induced transcriptomic responses at different axial locations of cortical bone would vary as a function of strain magnitude, we loaded the left tibias of 10-week-old female C57Bl/6 mice in vivo in compression, with contralateral limbs as controls. Animals were euthanized at 1, 3, or 24 hours post-loading or loaded for 1 week (n = 4-5/group). Bone marrow and cancellous bone were removed, cortical bone was segmented into the metaphyseal shell, proximal diaphysis, and mid-diaphysis, and load-induced differential gene expression and enriched biological processes were examined for the three segments. At each time point, the mid-diaphysis (highest strain) had the greatest transcriptomic response. Similarly, biological processes regulating bone formation and turnover increased earlier and to the greatest extent at the mid-diaphysis. Higher strain induced greater levels of osteoblast and osteocyte genes, whereas expression was lower in osteoclasts. Among the top differentially expressed genes at 24-hours post-loading, 17 had known functions in bone biology, of which 12 were present only in osteoblasts, 3 exclusively in osteoclasts, and 2 were present in both cell types. Based on these results, we conclude that murine tibial loading induces spatially unique transcriptomic responses correlating with strain magnitude in cortical bone. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Carolyn Chlebek
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jacob A Moore
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | | | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
7
|
Effects of Extracellular Osteoanabolic Agents on the Endogenous Response of Osteoblastic Cells. Cells 2021; 10:cells10092383. [PMID: 34572032 PMCID: PMC8471159 DOI: 10.3390/cells10092383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/07/2021] [Indexed: 12/27/2022] Open
Abstract
The complex multidimensional skeletal organization can adapt its structure in accordance with external contexts, demonstrating excellent self-renewal capacity. Thus, optimal extracellular environmental properties are critical for bone regeneration and inextricably linked to the mechanical and biological states of bone. It is interesting to note that the microstructure of bone depends not only on genetic determinants (which control the bone remodeling loop through autocrine and paracrine signals) but also, more importantly, on the continuous response of cells to external mechanical cues. In particular, bone cells sense mechanical signals such as shear, tensile, loading and vibration, and once activated, they react by regulating bone anabolism. Although several specific surrounding conditions needed for osteoblast cells to specifically augment bone formation have been empirically discovered, most of the underlying biomechanical cellular processes underneath remain largely unknown. Nevertheless, exogenous stimuli of endogenous osteogenesis can be applied to promote the mineral apposition rate, bone formation, bone mass and bone strength, as well as expediting fracture repair and bone regeneration. The following review summarizes the latest studies related to the proliferation and differentiation of osteoblastic cells, enhanced by mechanical forces or supplemental signaling factors (such as trace metals, nutraceuticals, vitamins and exosomes), providing a thorough overview of the exogenous osteogenic agents which can be exploited to modulate and influence the mechanically induced anabolism of bone. Furthermore, this review aims to discuss the emerging role of extracellular stimuli in skeletal metabolism as well as their potential roles and provide new perspectives for the treatment of bone disorders.
Collapse
|
8
|
Yang H, Bullock WA, Myhal A, DeShield P, Duffy D, Main RP. Cancellous Bone May Have a Greater Adaptive Strain Threshold Than Cortical Bone. JBMR Plus 2021; 5:e10489. [PMID: 33977205 PMCID: PMC8101616 DOI: 10.1002/jbm4.10489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/24/2021] [Accepted: 03/09/2021] [Indexed: 01/12/2023] Open
Abstract
Strain magnitude has a controlling influence on bone adaptive response. However, questions remain as to how and if cancellous and cortical bone tissues respond differently to varied strain magnitudes, particularly at a molecular level. The goal of this study was to characterize the time‐dependent gene expression, bone formation, and structural response of the cancellous and cortical bone of female C57Bl/6 mice to mechanical loading by applying varying load levels (low: −3.5 N; medium: −5.2 N; high: −7 N) to the skeleton using a mouse tibia loading model. The loading experiment showed that cortical bone mass at the tibial midshaft was significantly enhanced following all load levels examined and bone formation activities were particularly elevated at the medium and high loads applied. In contrast, for the proximal metaphyseal cancellous bone, only the high load led to significant increases in bone mass and bone formation indices. Similarly, expression of genes associated with inhibition of bone formation (e.g., Sost) was altered in the diaphyseal cortical bone at all load levels, but in the metaphyseal cortico‐cancellous bone only by the high load. Finite element analysis determined that the peak tensile or compressive strains that were osteogenic for the proximal cancellous bone under the high load were significantly greater than those that were osteogenic for the midshaft cortical tissues under the low load. These results suggest that the magnitude of the strain stimulus regulating structural, cellular, and molecular responses of bone to loading may be greater for the cancellous tissues than for the cortical tissues. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Haisheng Yang
- Department of Biomedical Engineering, Faculty of Environment and Life Beijing University of Technology Beijing China
| | | | - Alexandra Myhal
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences Purdue University West Lafayette IN USA
| | - Philip DeShield
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences Purdue University West Lafayette IN USA
| | - Daniel Duffy
- Weldon School of Biomedical Engineering Purdue University West Lafayette IN USA
| | - Russell P Main
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences Purdue University West Lafayette IN USA.,Weldon School of Biomedical Engineering Purdue University West Lafayette IN USA
| |
Collapse
|
9
|
Nepal AK, van Essen HW, van der Veen AJ, van Wieringen WN, Stavenuiter AWD, Cayami FK, Pals G, Micha D, Vanderschueren D, Lips P, Bravenboer N. Mechanical stress regulates bone regulatory gene expression independent of estrogen and vitamin D deficiency in rats. J Orthop Res 2021; 39:42-52. [PMID: 32530517 PMCID: PMC7818391 DOI: 10.1002/jor.24775] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 05/13/2020] [Accepted: 05/25/2020] [Indexed: 02/04/2023]
Abstract
Mechanical stress determines bone mass and structure. It is not known whether mechanical loading affects expression of bone regulatory genes in a combined deficiency of estrogen and vitamin D. We studied the effect of mechanical loading on the messenger RNA (mRNA) expression of bone regulatory genes during vitamin D and/or estrogen deficiency. We performed a single bout in vivo axial loading with 14 N peak load, 2 Hz frequency and 360 cycles in right ulnae of nineteen weeks old female control Wistar rats with or without ovariectomy (OVX), vitamin D deficiency and the combination of OVX and vitamin D deficiency (N = 10/group). Total bone RNA was isolated 6 hours after loading, and mRNA expression was detected of Mepe, Fgf23, Dmp1, Phex, Sost, Col1a1, Cyp27b1, Vdr, and Esr1. Serum levels of 25(OH)D, 1,25(OH)2 D and estradiol were also measured at this time point. The effect of loading, vitamin D and estrogen deficiency and their interaction on bone gene expression was tested using a mixed effect model analysis. Mechanical loading significantly increased the mRNA expression of Mepe, and Sost, whereas it decreased the mRNA expression of Fgf23 and Esr1. Mechanical loading showed a significant interaction with vitamin D deficiency with regard to mRNA expression of Vdr and Esr1. Mechanical loading affected gene expression of Mepe, Fgf23, Sost, and Esr1 independently of vitamin D or estrogen, indicating that mechanical loading may affect bone turnover even during vitamin D deficiency and after menopause.
Collapse
Affiliation(s)
- Ashwini Kumar Nepal
- Department of Clinical Chemistry, Amsterdam UMCVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Huib W. van Essen
- Department of Clinical Chemistry, Amsterdam UMCVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Albert J. van der Veen
- Department of Physics and Medical Technology, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Wessel N. van Wieringen
- Department of Epidemiology and Biostatistics, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of MathematicsVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Andrea W. D. Stavenuiter
- Department of Molecular Cell Biology and Immunology, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Ferdy Kurniawan Cayami
- Department of Clinical Genetics, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Anatomy, Center for Biomedical Research, Faculty of MedicineDiponegoro UniversitySemarangIndonesia
| | - Gerard Pals
- Department of Clinical Genetics, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Dirk Vanderschueren
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism, and AgeingKU LeuvenLeuvenBelgium
| | - Paul Lips
- Endocrine Section, Department of Internal Medicine, Amsterdam UMCVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam UMCVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
- Endocrine Section, Department of Internal Medicine, Amsterdam UMCVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
- Division Endocrinology, Department of Internal Medicine, Center of Bone QualityLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
10
|
Carina V, Della Bella E, Costa V, Bellavia D, Veronesi F, Cepollaro S, Fini M, Giavaresi G. Bone's Response to Mechanical Loading in Aging and Osteoporosis: Molecular Mechanisms. Calcif Tissue Int 2020; 107:301-318. [PMID: 32710266 DOI: 10.1007/s00223-020-00724-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Mechanotransduction is pivotal in the maintenance of homeostasis in different tissues and involves multiple cell signaling pathways. In bone, mechanical stimuli regulate the balance between bone formation and resorption; osteocytes play a central role in this regulation. Dysfunctions in mechanotransduction signaling or in osteocytes response lead to an imbalance in bone homeostasis. This alteration is very relevant in some conditions such as osteoporosis and aging. Both are characterized by increased bone weakness due to different causes, for example, the increase of osteocyte apoptosis that cause an alteration of fluid space, or the alteration of molecular pathways. There are intertwined yet very different mechanisms involved among the cell-intrinsic effects of aging on bone, the cell-intrinsic and tissue-level effects of estrogen/androgen withdrawal on bone, and the effects of reduced mechanical loading on bone, which are all involved to some degree in how aged bone fails to respond properly to stress/strain compared to younger bone. This review aims at clarifying how the cellular and molecular pathways regulated and induced in bone by mechanical stimulation are altered with aging and in osteoporosis, to highlight new possible targets for antiresorptive or anabolic bone therapeutic approaches.
Collapse
Affiliation(s)
- Valeria Carina
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy.
| | | | - Viviana Costa
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Daniele Bellavia
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Francesca Veronesi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Simona Cepollaro
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Milena Fini
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| |
Collapse
|
11
|
Main RP, Shefelbine SJ, Meakin LB, Silva MJ, van der Meulen MC, Willie BM. Murine Axial Compression Tibial Loading Model to Study Bone Mechanobiology: Implementing the Model and Reporting Results. J Orthop Res 2020; 38:233-252. [PMID: 31508836 PMCID: PMC9344861 DOI: 10.1002/jor.24466] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/23/2019] [Indexed: 02/04/2023]
Abstract
In vivo, tibial loading in mice is increasingly used to study bone adaptation and mechanotransduction. To achieve standardized and defined experimental conditions, loading parameters and animal-related factors must be considered when performing in vivo loading studies. In this review, we discuss these loading and animal-related experimental conditions, present methods to assess bone adaptation, and suggest reporting guidelines. This review originated from presentations by each of the authors at the workshop "Developing Best Practices for Mouse Models of In Vivo Loading" during the Preclinical Models Section at the Orthopaedic Research Society Annual Meeting, San Diego, CA, March 2017. Following the meeting, the authors engaged in detailed discussions with consideration of relevant literature. The guidelines and recommendations in this review are provided to help researchers perform in vivo loading experiments in mice, and thus further our knowledge of bone adaptation and the mechanisms involved in mechanotransduction. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:233-252, 2020.
Collapse
Affiliation(s)
- Russell P. Main
- Department of Basic Medical Sciences and Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA,Corresponding author: Russell Main ()
| | - Sandra J. Shefelbine
- Department of Bioengineering, Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, USA
| | - Lee B. Meakin
- Bristol Veterinary School, University of Bristol, Langford, Bristol BS40 5DU, UK
| | - Matthew J. Silva
- Departments of Orthopaedic Surgery and Biomedical Engineering, Musculoskeletal Research Center, Washington University, Saint Louis, MO, USA
| | - Marjolein C.H van der Meulen
- Meinig School of Biomedical Engineering and Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Bettina M. Willie
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| |
Collapse
|
12
|
Mechanobiological osteocyte feedback drives mechanostat regulation of bone in a multiscale computational model. Biomech Model Mechanobiol 2019; 18:1475-1496. [DOI: 10.1007/s10237-019-01158-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/23/2019] [Indexed: 10/26/2022]
|
13
|
Piet J, Hu D, Baron R, Shefelbine SJ. Bone adaptation compensates resorption when sciatic neurectomy is followed by low magnitude induced loading. Bone 2019; 120:487-494. [PMID: 30586636 DOI: 10.1016/j.bone.2018.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 01/02/2023]
Abstract
The uniaxial tibial loading model is commonly used to promote bone formation through mechanoadaptation in mice. Sciatic neurectomy on the other hand recruits osteoclasts, which results in bone loss. Previous studies have shown that combining sciatic neurectomy with high magnitude loading increases the amount of bone formed. Here we determine whether low-intensity loading (low magnitude and few cycles) is sufficient to maintain bone mass after sciatic neurectomy, either by promoting bone formation (balance between concurrent resorption and formation), or by preventing bone resorption altogether. We examined bone adaptation in 4 groups of female C57BL/6J mice, 19-22 weeks old: (1) sham surgery +10 N loading; (2) sham surgery +5 N loading; (3) sciatic neurectomy; (4) sciatic neurectomy +5 N loading. Left legs were kept intact as internal controls. We examined changes in bone cross sectional properties and marrow area with micro-CT images, and histomorphometric measures with histological sections at the midpoint between tibiofibular junctions. Loading at 10 N caused a significant increase in the amount of bone, but bone formation after 5 N of loading was not detectable in micro-CT images. There was significant bone loss in mice with sciatic neurectomy alone, but when combined with loading there was no significant bone loss. Histomorphometric analyses showed that loading at 5 N augmented bone formation periosteally on the lateral and posterior-medial surfaces, and reduced the number of endosteal osteoclasts on the posterior-medial surface compared to the contralateral leg. Combining sciatic neurectomy and loading at 5 N promoted faster mineral apposition on the periosteal lateral surface and augmented bone resorption on the endosteal posterior surface compared to the contralateral leg. These data demonstrate that low-intensity loading is sufficient to maintain bone mass after sciatic neurectomy, both by preventing recruitment of osteoclasts on the endosteal surface and by compensating endosteal resorption caused by disuse with periosteal formation promoted by loading. This has implications for the loading required to maintain bone mass after injury or prolonged bedrest.
Collapse
Affiliation(s)
- Judith Piet
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Dorothy Hu
- Department of Medicine, Harvard Medical School, Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Roland Baron
- Department of Medicine, Harvard Medical School, Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA; Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
14
|
NOTCH Signaling Is Activated through Mechanical Strain in Human Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells Int 2019; 2019:5150634. [PMID: 30936923 PMCID: PMC6413410 DOI: 10.1155/2019/5150634] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/28/2018] [Indexed: 11/18/2022] Open
Abstract
Skeletal development and remodeling of adult bone are critically controlled by activated NOTCH signaling in genetically modified mice. It is yet unclear whether NOTCH signaling is activated by mechanical strain sensed by bone cells. We found that expression of specific NOTCH target genes is induced after in vivo tibial mechanical loading in wild-type mice. We further applied mechanical strain through cyclic stretching in human bone marrow-derived mesenchymal stromal cells (BMSCs) in vitro by using a bioreactor system and detected upregulation of NOTCH target gene expression. Inhibition of the NOTCH pathway in primary BMSCs as well as telomerase-immortalized human BMSCs (hMSC-TERT) through the gamma-secretase inhibitor GSI XII blocked mechanotransduction and modulated actin cytoskeleton organization. Short-hairpin RNA gene silencing identified NOTCH2 as the key receptor mediating NOTCH effects on hMSC-TERT cells. Our data indicate a functional link between NOTCH activation and mechanotransduction in human BMSCs. We suggest that NOTCH signaling is an important contributor to molecular mechanisms that mediate the bone formation response to mechanical strain.
Collapse
|
15
|
The Bromodomain Inhibitor N-Methyl pyrrolidone Prevents Osteoporosis and BMP-Triggered Sclerostin Expression in Osteocytes. Int J Mol Sci 2018; 19:ijms19113332. [PMID: 30366476 PMCID: PMC6275050 DOI: 10.3390/ijms19113332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/09/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022] Open
Abstract
(1) Background: In an adult skeleton, bone is constantly renewed in a cycle of bone resorption, followed by bone formation. This coupling process, called bone remodeling, adjusts the quality and quantity of bone to the local needs. It is generally accepted that osteoporosis develops when bone resorption surpasses bone formation. Osteoclasts and osteoblasts, bone resorbing and bone forming cells respectively, are the major target in osteoporosis treatment. Inside bone and forming a complex network, the third and most abundant cells, the osteocytes, have long remained a mystery. Osteocytes are responsible for mechano-sensation and -transduction. Increased expression of the osteocyte-derived bone inhibitor sclerostin has been linked to estrogen deficiency-induced osteoporosis and is therefore a promising target for osteoporosis management. (2) Methods: Recently we showed in vitro and in vivo that NMP (N-Methyl-2-pyrrolidone) is a bioactive drug enhancing the BMP-2 (Bone Morphogenetic Protein 2) induced effect on bone formation while blocking bone resorption. Here we tested the effect of NMP on the expression of osteocyte-derived sclerostin. (3) Results: We found that NMP significantly decreased sclerostin mRNA and protein levels. In an animal model of osteoporosis, NMP prevented the estrogen deficiency-induced increased expression of sclerostin. (4) Conclusions: These results support the potential of NMP as a novel therapeutic compound for osteoporosis management, since it preserves bone by a direct interference with osteoblasts and osteoclasts and an indirect one via a decrease in sclerostin expression by osteocytes.
Collapse
|
16
|
Mikolajewicz N, Zimmermann EA, Willie BM, Komarova SV. Mechanically stimulated ATP release from murine bone cells is regulated by a balance of injury and repair. eLife 2018; 7:37812. [PMID: 30324907 PMCID: PMC6205812 DOI: 10.7554/elife.37812] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/28/2018] [Indexed: 02/06/2023] Open
Abstract
Bone cells sense and actively adapt to physical perturbations to prevent critical damage. ATP release is among the earliest cellular responses to mechanical stimulation. Mechanical stimulation of a single murine osteoblast led to the release of 70 ± 24 amole ATP, which stimulated calcium responses in neighboring cells. Osteoblasts contained ATP-rich vesicles that were released upon mechanical stimulation. Surprisingly, interventions that promoted vesicular release reduced ATP release, while inhibitors of vesicular release potentiated ATP release. Searching for an alternative ATP release route, we found that mechanical stresses induced reversible cell membrane injury in vitro and in vivo. Ca2+/PLC/PKC-dependent vesicular exocytosis facilitated membrane repair, thereby minimizing cell injury and reducing ATP release. Priming cellular repair machinery prior to mechanical stimulation reduced subsequent membrane injury and ATP release, linking cellular mechanosensitivity to prior mechanical exposure. Thus, our findings position ATP release as an integrated readout of membrane injury and repair. Athletes' skeletons get stronger with training, while bones weaken in people who cannot move or in astronauts experiencing weightlessness. This is because bone cells thrive when exposed to forces. When a bone cell is exposed to a physical force, the first thing that happens is the release of the energy-rich molecule called ATP into the space outside the cell. This molecule then binds to the neighboring cell to unleash a cascade of responses. ATP can exit the cell either through special canals in the cell membrane or released in tiny pod-like structures called vesicles. It is known that strong forces can injure the cell membrane and cause ATP to spill out. However, it is less clear how ATP is released when cells are subjected to regular forces. Mikolajewicz et al. investigated whether ATP exits through injured membranes of cells experiencing regular forces. Bone cells grown in the laboratory were gently poked with a glass needle or placed in a turbulent fluid to simulate forces experienced in the body. Dyes and fluorescent imaging techniques were used to observe the movement of vesicles and calculate the concentration of ATP in these cells. The experiments showed that regular forces in the body do indeed injure the cell membranes and cause ATP to spill out. But importantly, the cells repaired the injuries quickly by releasing vesicles that patch the wound. As soon as the membrane is sealed, ATP stops coming out. From the first injury, cells adapted and quickly strengthened their membrane and repair system to be more resilient against future forces. This process was also seen in the shin bones of mice. These results are important because knowing how bone cells sense, respond and convert physical forces can help us develop treatments for astronauts, the injured and aged.
Collapse
Affiliation(s)
- Nicholas Mikolajewicz
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| | - Elizabeth A Zimmermann
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Department of Pediatric Surgery, Montreal, Quebec, Canada
| | - Bettina M Willie
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Department of Pediatric Surgery, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Tajima T, Menuki K, Okuma KF, Tsukamoto M, Fukuda H, Okada Y, Kosugi K, Yamanaka Y, Uchida S, Sakai A. Cortical bone loss due to skeletal unloading in aldehyde dehydrogenase 2 gene knockout mice is associated with decreased PTH receptor expression in osteocytes. Bone 2018; 110:254-266. [PMID: 29482068 DOI: 10.1016/j.bone.2018.02.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/09/2018] [Accepted: 02/21/2018] [Indexed: 01/22/2023]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is the enzyme that degrades and detoxifies the acetaldehyde produced by alcohol metabolism. In our previous study, we found that compared with wild-type mice (WT), climbing exercises did not increase trabecular bone mass in Aldh2 knockout mice (KO). The purpose of this study was to clarify the effect of the Aldh2 gene on cortical bone structure and on the dynamics of skeletal unloading. Eight-week-old male KO and WT were divided into ground control (GC) or tail suspension (TS) groups for one week (i.e., the KOGC, KOTS, WTGC and WTTS groups). We measured the bone mineral density (BMD) of the femur using dual-energy X-ray absorptiometry. We assessed the femoral morphometry using peripheral quantitative computed tomography (pQCT) and evaluated the femoral cortex histomorphometry, and cortical mRNA using quantitative RT-PCR and cortical bone immunohistostaining. No significant differences were found between the femoral BMD of WTGC and that of WTTS, but the BMD in KOTS was significantly lower than that of KOGC. The pQCT results revealed that the cortical BMD of the femoral diaphysis in KOTS was significantly lower than that of KOGC. Furthermore, the cortical bone area and cortical thickness were significantly lower in KOTS than in the other three groups. Cortical histomorphometric analysis revealed that the endosteal and periosteal bone formation parameters were significantly lower in KOTS than in KOGC. Bone formation signals such as parathyroid hormone receptor (PTHR) were significantly decreased in KOTS compared with the levels in KOGC. Cortical bone immunohistostaining revealed a significantly decreased expression of PTHR in the osteocytes of KOTS compared with the expression level in KOGC. Thus, we concluded that when the Aldh2 gene is disrupted, skeletal unloading suppresses bone formation to decrease cortical bone mass, which may be mediated by a decreased expression of PTH receptors in osteocytes.
Collapse
Affiliation(s)
- Takafumi Tajima
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kunitaka Menuki
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Kayoko Furukawa Okuma
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Manabu Tsukamoto
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Hokuto Fukuda
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yasuaki Okada
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kenji Kosugi
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiaki Yamanaka
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Soshi Uchida
- Department of Orthpaedic Surgery, Wakamatsu Hospital for the University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Akinori Sakai
- Department of Orthpaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| |
Collapse
|
18
|
Haffner-Luntzer M, Kovtun A, Lackner I, Mödinger Y, Hacker S, Liedert A, Tuckermann J, Ignatius A. Estrogen receptor α- (ERα), but not ERβ-signaling, is crucially involved in mechanostimulation of bone fracture healing by whole-body vibration. Bone 2018; 110:11-20. [PMID: 29367057 DOI: 10.1016/j.bone.2018.01.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/21/2017] [Accepted: 01/14/2018] [Indexed: 02/07/2023]
Abstract
Mechanostimulation by low-magnitude high frequency vibration (LMHFV) has been shown to provoke anabolic effects on the intact skeleton in both mice and humans. However, experimental studies revealed that, during bone fracture healing, the effect of whole-body vibration is profoundly influenced by the estrogen status. LMHFV significantly improved fracture healing in ovariectomized (OVX) mice being estrogen deficient, whereas bone regeneration was significantly reduced in non-OVX, estrogen-competent mice. Furthermore, estrogen receptors α (ERα) and β (ERβ) were differentially expressed in the fracture callus after whole-body vibration, depending on the estrogen status. Based on these data, we hypothesized that ERs may mediate vibration-induced effects on fracture healing. To prove this hypothesis, we investigated the effects of LMHFV on bone healing in mice lacking ERα or ERβ. To study the influence of the ER ligand estrogen, both non-OVX and OVX mice were used. All mice received a femur osteotomy stabilized by an external fixator. Half of the mice were sham-operated or subjected to OVX 4 weeks before osteotomy. Half of each group received LMHFV with 0.3 g and 45 Hz for 20 min per day, 5 days per week. After 21 days, fracture healing was evaluated by biomechanical testing, μCT analysis, histomorphometry and immunohistochemistry. Absence of ERα or ERβ did not affect fracture healing in sham-treated mice. Wildtype (WT) and ERβ-knockout mice similarly displayed impaired bone regeneration after OVX, whereas ERα-knockout mice did not. Confirming previous data, in WT mice, LMHFV negatively affected bone repair in non-OVX mice, whereas OVX-induced compromised healing was significantly improved by vibration. In contrast, vibrated ERα-knockout mice did not display significant differences in fracture healing compared to non-vibrated animals, both in non-OVX and OVX mice. Fracture healing in ERβ-knockout mice was similarly affected by LMHFV as in WT mice. These results suggest that ERα-signaling may be crucial for vibration-induced effects on fracture healing, whereas ERβ-signaling may play a minor role.
Collapse
Affiliation(s)
- Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany.
| | - Anna Kovtun
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| | - Ina Lackner
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| | - Yvonne Mödinger
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| | - Steffen Hacker
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstraße 8, 89081 Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| |
Collapse
|
19
|
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: 3.3] [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
|
20
|
Recker RR, Lappe JM, Davies M, Kimmel D. Perimenopausal bone histomorphometry before and after menopause. Bone 2018; 108:55-61. [PMID: 29258873 DOI: 10.1016/j.bone.2017.12.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 01/18/2023]
Abstract
Investigators and clinicians have had few normal bone histomorphometry data available to compare with those found in diseased patients, or in the results of treatments. The Goals and Objectives of this work are two-fold: 1. to present static and dynamic bone histomorphometry data from transilial bone biopsies performed on 76 healthy, premenopausal women. 2. To present paired static and dynamic bone histomorphometry data from bone biopsies on a subset (N=51 pairs) of these same healthy women whose biopsies were repeated 12months after their last menses. Statistical comparisons between the pre- and postmenopausal data are presented. These data will shrink this important gap, both for clinicians and investigators. We enrolled 76 healthy, premenopausal women over age 46, performed transilial bone biopsies after tetracycline labeling, and during a period of 9.5years, we re-biopsied 51 of them who passed through menopause and remained healthy the entire time. We also obtained serum biochemical measurements, and serial DXA exams during the period of observation. The dynamic bone histomorphometry demonstrated a doubling of bone remodeling, and increases in serum bone markers at the time of the second biopsy. Lumbar spine bone density also declined, and there were significant correlations between serum markers and histomorphometry variables. The data demonstrate that healthy menopause results in an important increase in bone remodeling, and a loss of bone density. We do not fully understand the mechanisms of these transmenopausal changes, but the data provide some clues that are helpful.
Collapse
Affiliation(s)
- Robert R Recker
- Creighton University School of Medicine, 6829 N 72nd St, Ste 7400, Omaha, NE, 68122, United States.
| | - Joan M Lappe
- Creighton University School of Medicine, 6829 N 72nd St, Ste 7400, Omaha, NE, 68122, United States.
| | - Michael Davies
- Creighton University, 6829 N 72nd St, Ste 7400, Omaha, NE, 68122, United States.
| | - Donald Kimmel
- Creighton University School of Medicine, 6829 N 72nd St, Ste 7400, Omaha, NE, 68122, United States
| |
Collapse
|
21
|
Scheuren A, Wehrle E, Flohr F, Müller R. Bone mechanobiology in mice: toward single-cell in vivo mechanomics. Biomech Model Mechanobiol 2017; 16:2017-2034. [PMID: 28735414 DOI: 10.1007/s10237-017-0935-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 07/11/2017] [Indexed: 01/27/2023]
Abstract
Mechanically driven bone (re)modeling is a multiscale process mediated through complex interactions between multiple cell types and their microenvironments. However, the underlying mechanisms of how cells respond to mechanical signals are still unclear and are at the focus of the field of bone mechanobiology. Traditionally, this complex process has been addressed by reducing the system to single scales and cell types. It is only recently that more integrative approaches have been established to study bone mechanobiology across multiple scales in which mechanical load at the organ level is related to molecular responses at the cellular level. The availability of mouse loading models and imaging techniques with improved spatial and temporal resolution has made it possible to track dynamic bone (re)modeling at the tissue and cellular level in vivo. Coupled with advanced computational models, the (re)modeling activities at the tissue scale can be associated with the mechanical microenvironment. However, methods are lacking to link the molecular responses of different cell types to their local mechanical microenvironment and bone (re)modeling activities occurring at the tissue scale. With recent improvements in "omics" technologies and single-cell molecular biology, it is now possible to sequence the complete genome and transcriptome of single cells. These technologies offer unique opportunities to comprehensively investigate the cellular transcriptional profiles within their specific microenvironment. By combining single-cell "omics" technologies with well-established tissue-scale models of bone mechanobiology, we propose a mechanomics approach to locally analyze the transcriptome of single cells with respect to their local 3D mechanical in vivo environment.
Collapse
Affiliation(s)
- Ariane Scheuren
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Felicitas Flohr
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland.
| |
Collapse
|
22
|
Galea GL, Lanyon LE, Price JS. Sclerostin's role in bone's adaptive response to mechanical loading. Bone 2017; 96:38-44. [PMID: 27742499 PMCID: PMC5340132 DOI: 10.1016/j.bone.2016.10.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/27/2016] [Accepted: 10/10/2016] [Indexed: 01/08/2023]
Abstract
Mechanical loading is the primary functional determinant of bone mass and architecture, and osteocytes play a key role in translating mechanical signals into (re)modelling responses. Although the precise mechanisms remain unclear, Wnt signalling pathway components, and the anti-osteogenic canonical Wnt inhibitor Sost/sclerostin in particular, play an important role in regulating bone's adaptive response to loading. Increases in loading-engendered strains down-regulate osteocyte sclerostin expression, whereas reduced strains, as in disuse, are associated with increased sclerostin production and bone loss. However, while sclerostin up-regulation appears to be necessary for the loss of bone with disuse, the role of sclerostin in the osteogenic response to loading is more complex. While mice unable to down-regulate sclerostin do not gain bone with loading, Sost knockout mice have an enhanced osteogenic response to loading. The molecular mechanisms by which osteocytes sense and transduce loading-related stimuli into changes in sclerostin expression remain unclear but include several, potentially interlinked, signalling cascades involving periostin/integrin, prostaglandin, estrogen receptor, calcium/NO and Igf signalling. Deciphering the mechanisms by which changes in the mechanical environment regulate sclerostin production may lead to the development of therapeutic strategies that can reverse the skeletal structural deterioration characteristic of disuse and age-related osteoporosis and enhance bones' functional adaptation to loading. By enhancing the osteogenic potential of the context in which individual therapies such as sclerostin antibodies act it may become possible to both prevent and reverse the age-related skeletal structural deterioration characteristic of osteoporosis.
Collapse
Affiliation(s)
- Gabriel L Galea
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London WC1N 1EH, United Kingdom; School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU, United Kingdom.
| | - Lance E Lanyon
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU, United Kingdom
| | - Joanna S Price
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU, United Kingdom
| |
Collapse
|
23
|
Galea GL, Meakin LB, Harris MA, Delisser PJ, Lanyon LE, Harris SE, Price JS. Old age and the associated impairment of bones' adaptation to loading are associated with transcriptomic changes in cellular metabolism, cell-matrix interactions and the cell cycle. Gene 2017; 599:36-52. [PMID: 27840164 PMCID: PMC5139832 DOI: 10.1016/j.gene.2016.11.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 11/06/2016] [Indexed: 02/08/2023]
Abstract
In old animals, bone's ability to adapt its mass and architecture to functional load-bearing requirements is diminished, resulting in bone loss characteristic of osteoporosis. Here we investigate transcriptomic changes associated with this impaired adaptive response. Young adult (19-week-old) and aged (19-month-old) female mice were subjected to unilateral axial tibial loading and their cortical shells harvested for microarray analysis between 1h and 24h following loading (36 mice per age group, 6 mice per loading group at 6 time points). In non-loaded aged bones, down-regulated genes are enriched for MAPK, Wnt and cell cycle components, including E2F1. E2F1 is the transcription factor most closely associated with genes down-regulated by ageing and is down-regulated at the protein level in osteocytes. Genes up-regulated in aged bone are enriched for carbohydrate metabolism, TNFα and TGFβ superfamily components. Loading stimulates rapid and sustained transcriptional responses in both age groups. However, genes related to proliferation are predominantly up-regulated in the young and down-regulated in the aged following loading, whereas those implicated in bioenergetics are down-regulated in the young and up-regulated in the aged. Networks of inter-related transcription factors regulated by E2F1 are loading-responsive in both age groups. Loading regulates genes involved in similar signalling cascades in both age groups, but these responses are more sustained in the young than aged. From this we conclude that cells in aged bone retain the capability to sense and transduce loading-related stimuli, but their ability to translate acute responses into functionally relevant outcomes is diminished.
Collapse
Affiliation(s)
- Gabriel L Galea
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Lee B Meakin
- School of Veterinary Sciences, University of Bristol, Bristol, UK.
| | - Marie A Harris
- Department of Periodontics & Cellular and Structural Biology, University of Texas Health Science Centre, San Antonio, USA
| | - Peter J Delisser
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Lance E Lanyon
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Stephen E Harris
- Department of Periodontics & Cellular and Structural Biology, University of Texas Health Science Centre, San Antonio, USA
| | - Joanna S Price
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
24
|
Holguin N, Brodt MD, Silva MJ. Activation of Wnt Signaling by Mechanical Loading Is Impaired in the Bone of Old Mice. J Bone Miner Res 2016; 31:2215-2226. [PMID: 27357062 PMCID: PMC5397287 DOI: 10.1002/jbmr.2900] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 01/18/2023]
Abstract
Aging diminishes bone formation engendered by mechanical loads, but the mechanism for this impairment remains unclear. Because Wnt signaling is required for optimal loading-induced bone formation, we hypothesized that aging impairs the load-induced activation of Wnt signaling. We analyzed dynamic histomorphometry of 5-month-old, 12-month-old, and 22-month-old C57Bl/6JN mice subjected to multiple days of tibial compression and corroborated an age-related decline in the periosteal loading response on day 5. Similarly, 1 day of loading increased periosteal and endocortical bone formation in young-adult (5-month-old) mice, but old (22-month-old) mice were unresponsive. These findings corroborated mRNA expression of genes related to bone formation and the Wnt pathway in tibias after loading. Multiple bouts (3 to 5 days) of loading upregulated bone formation-related genes, e.g., Osx and Col1a1, but older mice were significantly less responsive. Expression of Wnt negative regulators, Sost and Dkk1, was suppressed with a single day of loading in all mice, but suppression was sustained only in young-adult mice. Moreover, multiple days of loading repeatedly suppressed Sost and Dkk1 in young-adult, but not in old tibias. The age-dependent response to loading was further assessed by osteocyte staining for Sclerostin and LacZ in tibia of TOPGAL mice. After 1 day of loading, fewer osteocytes were Sclerostin-positive and, corroboratively, more osteocytes were LacZ-positive (Wnt active) in both 5-month-old and 12-month-old mice. However, although these changes were sustained after multiple days of loading in 5-month-old mice, they were not sustained in 12-month-old mice. Last, Wnt1 and Wnt7b were the most load-responsive of the 19 Wnt ligands. However, 4 hours after a single bout of loading, although their expression was upregulated threefold to 10-fold in young-adult mice, it was not altered in old mice. In conclusion, the reduced bone formation response of aged mice to loading may be due to failure to sustain Wnt activity with repeated loading. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nilsson Holguin
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, USA
| | - Michael D Brodt
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| |
Collapse
|
25
|
Liakou CG, Mastorakos G, Makris K, Fatouros IG, Avloniti A, Marketos H, Antoniou JD, Galanos A, Dontas I, Rizos D, Tournis S. Changes of serum sclerostin and Dickkopf-1 levels during the menstrual cycle. A pilot study. Endocrine 2016; 54:543-551. [PMID: 27601021 DOI: 10.1007/s12020-016-1056-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/06/2016] [Indexed: 12/17/2022]
Abstract
Studies in postmenopausal women have identified sclerostin as a strong candidate for mediating estrogen effects on the skeleton. The effects of estradiol on sclerostin and Dickkopf-1 in younger women remain unclear. The main purpose of this study is to investigate the impact of estradiol and gonadotrophins fluctuations during the menstrual cycle on circulating sclerostin and Dickkopf-1 levels and the possible relationship of sclerostin and Dickkopf-1 with changes in N-terminal propeptide of type 1 collagen and C-telopeptide of collagen cross-links. Fourteen healthy premenopausal Caucasian women, with regular menses, aged 33.6 ± 4.5 years participated. After the first day of menstruation and every-other-day up to the next menses, fasting serum estradiol, luteinizing hormone, follicle-stimulating hormone, sclerostin, Dickkopf-1, N-terminal propeptide of type 1 collagen, and C-telopeptide of collagen cross-links levels were measured in peripheral blood. Participants completed dietary questionnaires and the International physical activity questionnaire during the cycle. Neither sclerostin nor Dickkopf-1 levels changed significantly across the menstrual cycle (p = 0.18 and p = 0.39, respectively), while N-terminal propeptide of type 1 collagen and C-telopeptide of collagen cross-links levels presented cyclic variation (p < 0.001 and p = 0.004, respectively). Baseline sclerostin (29.23 ± 10.62 pmol/L) positively correlated with N-terminal propeptide of type 1 collagen (r = 0.71, p < 0.01) and C-telopeptide of collagen cross-links (r = 0.63, p < 0.05), while Dickkopf-1 (4.82 ± 2.23 pmol/L) correlated positively with N-terminal propeptide of type 1 collagen (r = 0.56, p < 0.05). Mid-cycle E2 levels presented significant negative association with the percent decrease of C-telopeptide of collagen cross-links at all-time points during the luteal period (r = -0.60 to -0.68, p < 0.05-0.01). Circulating sclerostin and Dickkopf-1 levels do not change across the menstrual cycle and do not demonstrate any relationship with estradiol in premenopausal women. Further investigation is needed concerning the role of sclerostin and Dickkopf-1 on bone turnover in young estrogen-sufficient women.
Collapse
Affiliation(s)
- Chrysoula G Liakou
- Laboratory for Research of the Musculoskeletal System "Th. Garofalidis", Medical School, Athens University, KAT Hospital, 10 Athinas Str., Kifissia, 14561, Athens, Greece.
| | - George Mastorakos
- Unit of Endocrinology, Diabetes and Metabolism, Aretaieion Hospital, Athens Medical School, Athens University, 76 Vas. Sofias Av., 11528, Athens, Greece
| | - Konstantinos Makris
- Department of Biochemistry, KAT Hospital, 10 Athinas Str., Kifissia, 14561, Athens, Greece
| | - Ioannis G Fatouros
- School of Physical Education and Sports Sciences, University of Thessaly, 42100, Karies, Trikala, Greece
| | - Alexandra Avloniti
- School of Physical Education and Sports Sciences, Democritus University of Thrace, 69100, Komotini, Greece
| | - Helen Marketos
- Department of Biochemistry, KAT Hospital, 10 Athinas Str., Kifissia, 14561, Athens, Greece
| | - Julia D Antoniou
- Laboratory for Research of the Musculoskeletal System "Th. Garofalidis", Medical School, Athens University, KAT Hospital, 10 Athinas Str., Kifissia, 14561, Athens, Greece
| | - Antonios Galanos
- Laboratory for Research of the Musculoskeletal System "Th. Garofalidis", Medical School, Athens University, KAT Hospital, 10 Athinas Str., Kifissia, 14561, Athens, Greece
| | - Ismene Dontas
- Laboratory for Research of the Musculoskeletal System "Th. Garofalidis", Medical School, Athens University, KAT Hospital, 10 Athinas Str., Kifissia, 14561, Athens, Greece
| | - Demetrios Rizos
- Hormone Laboratory, Aretaieion Hospital, Medical School, Athens University, 76 Vas. Sofias Av., 11528, Athens, Greece
| | - Symeon Tournis
- Laboratory for Research of the Musculoskeletal System "Th. Garofalidis", Medical School, Athens University, KAT Hospital, 10 Athinas Str., Kifissia, 14561, Athens, Greece
| |
Collapse
|
26
|
Al-Kattan R, Retzepi M, Calciolari E, Donos N. Microarray gene expression during early healing of GBR-treated calvarial critical size defects. Clin Oral Implants Res 2016; 28:1248-1257. [PMID: 27616585 DOI: 10.1111/clr.12949] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVES To investigate the gene expression and molecular pathways implicated in the regulation of the osseous healing process following guided bone regeneration (GBR). MATERIAL AND METHODS Six 6-month-old Wistar male rats were used. Standardized 5-mm critical size defects were created in the parietal bones of each animal and treated with an extracranial and intracranial ePTFE membrane, according to the GBR principle. Three animals were randomly sacrificed after 7 and 15 days of healing. Total RNA was extracted from each sample and prepared for gene expression analysis. RNA quality and quantity were assessed, followed by hybridization of the cRNA to Affymetrix GeneChip Rat Genome 230 2.0 Arrays. The Affymetrix data were processed, and first-order analysis, quality control and statistical analysis were performed. Biological interpretation was performed via pathway and Gene Ontology (GO) analysis. RESULTS Between the 7- and 15-day samples, 538 genes were differently regulated. At day 7, inflammatory and immune responses were clearly upregulated. In addition, GO terms related to angiogenesis and cell cycle regulation were overexpressed. At day 15, a more complex cellular activity and cell metabolism were evident. The bone formation processes were significantly overexpressed, with several genes encoding growth factors, enzyme activity, and extracellular matrix formation found as upregulated. Remarkably, a negative regulation of Wnt signalling pathway was observed at 15 days. DISCUSSION The gene expression profile of the cells participating in osseous formation varied depending on the healing stage. A number of candidate genes that seem differentially expressed during early stages of intramembranous bone regeneration was suggested.
Collapse
Affiliation(s)
- R Al-Kattan
- Department of Periodontics and Community Dentistry, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | | | - E Calciolari
- Department of Periodontology, UCL Eastman Dental Institute, London, UK.,Centre for Oral Clinical Research, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London (QMUL), London, UK
| | - N Donos
- Centre for Oral Clinical Research, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London (QMUL), London, UK
| |
Collapse
|
27
|
The molecular basis of bone mechanotransduction. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2016; 16:221-36. [PMID: 27609037 PMCID: PMC5114345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The skeleton has the ability to perfectly adapt to external forces of the operating environment, by altering its morphology and metabolism in order to meet different needs. This unique adaptive capacity of the skeleton creates an interesting range of biological questions concerning the perception of mechanical or other kinds of signals, the type of receptor, and the molecular pathways involved in this adaptation. Studies of the characteristics of the cellular engineering provide a host of new information that confers to osteocytes the role of the protagonist in the perception and regulation of mechanical effects on the skeleton. The identity of mechanoreceptors is manifold and concerns ion channels, integrins, cell membrane, the cytoskeleton, and other systems. A similar multiplicity characterizes the intracellular signaling. This review describes recent data concerning the outward force reception systems and intracellular transduction pathways of information transfer leading to the continuous adaptation of bone tissue. Increased appreciation of the importance of the mechanical environment in regulating and determining the effectiveness of structural adjustment of the skeleton defines new horizons for the discovery of novel therapeutic approaches to diseases associated with bone loss.
Collapse
|
28
|
Kelly NH, Schimenti JC, Ross FP, van der Meulen MCH. Transcriptional profiling of cortical versus cancellous bone from mechanically-loaded murine tibiae reveals differential gene expression. Bone 2016; 86:22-9. [PMID: 26876048 PMCID: PMC4833881 DOI: 10.1016/j.bone.2016.02.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/10/2016] [Accepted: 02/11/2016] [Indexed: 12/19/2022]
Abstract
Mechanical loading is an anabolic stimulus that increases bone mass, and thus a promising method to counteract osteoporosis-related bone loss. The mechanism of this anabolism remains unclear, and needs to be established for both cortical and cancellous envelopes individually. We hypothesized that cortical and cancellous bone display different gene expression profiles at baseline and in response to mechanical loading. To test this hypothesis, the left tibiae of 10-week-old female C57Bl/6 mice were subjected to one session of axial tibial compression (9N, 1200cycles, 4Hz triangle waveform) and euthanized 3 and 24h following loading. The right limb served as the contralateral control. We performed RNA-seq on marrow-free metaphyseal samples from the cortical shell and the cancellous core to determine differential gene expression at baseline (control limb) and in response to load. Differential expression was verified with qPCR. Cortical and cancellous bone exhibited distinctly different transcriptional profiles basally and in response to mechanical loading. More genes were differentially expressed with loading at 24h with more genes downregulated at 24h than at 3h in both tissues. Enhanced Wnt signaling dominated the response in cortical bone at 3 and 24h, but in cancellous bone only at 3h. In cancellous bone at 24h many muscle-related genes were downregulated. These findings reveal key differences between cortical and cancellous genetic regulation in response to mechanical loading. Future studies at different time points and multiple loading sessions will add to our knowledge of cortical and cancellous mechanotransduction with the potential to identify new targets for mouse genetic knockout studies and drugs to treat osteoporosis.
Collapse
Affiliation(s)
- Natalie H Kelly
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, 105 Upson Hall, Ithaca, NY 14853, USA; Nancy E and Peter C Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, Ithaca, NY 14853, USA.
| | - John C Schimenti
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - F Patrick Ross
- Research Division, Hospital for Special Surgery, 541 East 71st St., New York, NY 10021, USA.
| | - Marjolein C H van der Meulen
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, 105 Upson Hall, Ithaca, NY 14853, USA; Nancy E and Peter C Meinig School of Biomedical Engineering, Cornell University, 101 Weill Hall, Ithaca, NY 14853, USA; Research Division, Hospital for Special Surgery, 541 East 71st St., New York, NY 10021, USA.
| |
Collapse
|
29
|
Navein AE, Cooke EJ, Davies JR, Smith TG, Wells LHM, Ohazama A, Healy C, Sharpe PT, Evans SL, Evans BAJ, Votruba M, Wells T. Disrupted mitochondrial function in the Opa3L122P mouse model for Costeff Syndrome impairs skeletal integrity. Hum Mol Genet 2016; 25:2404-2416. [PMID: 27106103 PMCID: PMC5181626 DOI: 10.1093/hmg/ddw107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction connects metabolic disturbance with numerous pathologies, but the significance of mitochondrial activity in bone remains unclear. We have, therefore, characterized the skeletal phenotype in the Opa3L122P mouse model for Costeff syndrome, in which a missense mutation of the mitochondrial membrane protein, Opa3, impairs mitochondrial activity resulting in visual and metabolic dysfunction. Although widely expressed in the developing normal mouse head, Opa3 expression was restricted after E14.5 to the retina, brain, teeth and mandibular bone. Opa3 was also expressed in adult tibiae, including at the trabecular surfaces and in cortical osteocytes, epiphyseal chondrocytes, marrow adipocytes and mesenchymal stem cell rosettes. Opa3L122P mice displayed craniofacial abnormalities, including undergrowth of the lower mandible, accompanied in some individuals by cranial asymmetry and incisor malocclusion. Opa3L122P mice showed an 8-fold elevation in tibial marrow adiposity, due largely to increased adipogenesis. In addition, femoral length and cortical diameter and wall thickness were reduced, the weakening of the calcified tissue and the geometric component of strength reducing overall cortical strength in Opa3L122P mice by 65%. In lumbar vertebrae reduced vertebral body area and wall thickness were accompanied by a proportionate reduction in marrow adiposity. Although the total biomechanical strength of lumbar vertebrae was reduced by 35%, the strength of the calcified tissue (σmax) was proportionate to a 38% increase in trabecular number. Thus, mitochondrial function is important for the development and maintenance of skeletal integrity, impaired bone growth and strength, particularly in limb bones, representing a significant new feature of the Costeff syndrome phenotype.
Collapse
Affiliation(s)
- Alice E Navein
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Esther J Cooke
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Jennifer R Davies
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4LU, UK
| | - Terence G Smith
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4LU, UK
| | - Lois H M Wells
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.,Caerleon Comprehensive School, Caerleon, Newport NP18 1NF, UK
| | - Atsushi Ohazama
- Department of Craniofacial Development and Stem Cell Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Christopher Healy
- Department of Craniofacial Development and Stem Cell Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Paul T Sharpe
- Department of Craniofacial Development and Stem Cell Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Sam L Evans
- School of Engineering, Cardiff University, The Parade, Cardiff CF24 3AA, UK
| | - Bronwen A J Evans
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4LU, UK.,Cardiff Eye Unit, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Timothy Wells
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| |
Collapse
|
30
|
Abstract
Mechanical loading is a potent anabolic regulator of bone mass, and the first line of defense for bone loss is weight-bearing exercise. Likewise, protected weight bearing is the first prescribed physical therapy following orthopedic reconstructive surgery. In both cases, enhancement of new bone formation is the goal. Our understanding of the physical cues, mechanisms of force sensation, and the subsequent cellular response will help identify novel physical and therapeutic treatments for age- and disuse-related bone loss, delayed- and nonunion fractures, and significant bony defects. This review highlights important new insights into the principles and mechanisms governing mechanical adaptation of the skeleton during homeostasis and repair and ends with a summary of clinical implications stemming from our current understanding of how bone adapts to biophysical force.
Collapse
|
31
|
Meakin LB, Delisser PJ, Galea GL, Lanyon LE, Price JS. Disuse rescues the age-impaired adaptive response to external loading in mice. Osteoporos Int 2015; 26:2703-8. [PMID: 25920749 PMCID: PMC4605986 DOI: 10.1007/s00198-015-3142-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/17/2015] [Indexed: 01/06/2023]
Abstract
UNLABELLED We aimed to determine whether aged bone's diminished response to mechanical loading could be rescued by modulating habitual activity. By reducing background loading, aged bone's response to loading increased to a level no different to young mice. This suggests, given the right stimulus, that ageing bone can respond to mechanical loading. INTRODUCTION Age-related decline in bone mass has been suggested to represent an impaired ability of bone to adapt to its mechanical environment. In young mice, the tibia's response to external mechanical loading has been shown to increase when habitual activity is reduced by sciatic neurectomy. Here we investigate if neurectomy can rescue bone's response to loading in old mice. METHODS The effect of tibial disuse, induced by unilateral sciatic neurectomy (SN), on the adaptive response to a single peak magnitude of dynamic load-engendered mechanical strain was assessed in 19-month-old (aged) mice. In a second experiment, a range of peak loads was used to assess the load magnitude-related effects of loading on a background of disuse in young adult and aged mice. Bone architecture was analysed using micro-computed tomography (μCT) and dynamic histomorphometry. RESULTS In the first experiment, SN in aged mice was associated with a significant periosteal osteogenic response to loading not observed in sham-operated mice (7.98 ± 1.7 vs 1.02 ± 2.2 % increase in periosteally enclosed area, p < 0.05). In the second experiment, SN abrogated the expected age-related difference in the bones' osteogenic response to peak strain magnitude (p > 0.05). CONCLUSIONS These data suggest that bones' age-related decline in osteogenic responsiveness to loading does not originate in bone cells to either assess, or appropriately respond to strain, but rather is likely to be due to inhibitory "averaging" effects derived from the habitual strains to which the bone is already adapted. If such "strain averaging" is applicable to humans, it suggests that gentle exercise may degrade the beneficially osteogenic effects of short periods of more vigorous activity.
Collapse
Affiliation(s)
- L B Meakin
- School of Veterinary Science, University of Bristol, Langford, Bristol, BS40 5DU, UK.
| | - P J Delisser
- School of Veterinary Science, University of Bristol, Langford, Bristol, BS40 5DU, UK
| | - G L Galea
- School of Veterinary Science, University of Bristol, Langford, Bristol, BS40 5DU, UK
| | - L E Lanyon
- School of Veterinary Science, University of Bristol, Langford, Bristol, BS40 5DU, UK
| | - J S Price
- School of Veterinary Science, University of Bristol, Langford, Bristol, BS40 5DU, UK
| |
Collapse
|
32
|
Shea CA, Rolfe RA, Murphy P. The importance of foetal movement for co-ordinated cartilage and bone development in utero : clinical consequences and potential for therapy. Bone Joint Res 2015; 4:105-16. [PMID: 26142413 PMCID: PMC4602203 DOI: 10.1302/2046-3758.47.2000387] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Construction of a functional skeleton is accomplished
through co-ordination of the developmental processes of chondrogenesis,
osteogenesis, and synovial joint formation. Infants whose movement in
utero is reduced or restricted and who subsequently suffer
from joint dysplasia (including joint contractures) and thin hypo-mineralised
bones, demonstrate that embryonic movement is crucial for appropriate
skeletogenesis. This has been confirmed in mouse, chick, and zebrafish
animal models, where reduced or eliminated movement consistently yields
similar malformations and which provide the possibility of experimentation
to uncover the precise disturbances and the mechanisms by which
movement impacts molecular regulation. Molecular genetic studies have
shown the important roles played by cell communication signalling
pathways, namely Wnt, Hedgehog, and transforming growth factor-beta/bone
morphogenetic protein. These pathways regulate cell behaviours such
as proliferation and differentiation to control maturation of the
skeletal elements, and are affected when movement is altered. Cell
contacts to the extra-cellular matrix as well as the cytoskeleton
offer a means of mechanotransduction which could integrate mechanical
cues with genetic regulation. Indeed, expression of cytoskeletal
genes has been shown to be affected by immobilisation. In addition
to furthering our understanding of a fundamental aspect of cell control
and differentiation during development, research in this area is
applicable to the engineering of stable skeletal tissues from stem
cells, which relies on an understanding of developmental mechanisms
including genetic and physical criteria. A deeper understanding
of how movement affects skeletogenesis therefore has broader implications
for regenerative therapeutics for injury or disease, as well as
for optimisation of physical therapy regimes for individuals affected
by skeletal abnormalities. Cite this article: Bone Joint Res 2015;4:105–116
Collapse
Affiliation(s)
- C A Shea
- Trinity College Dublin, College Green, Dublin, D2, Ireland
| | | | - P Murphy
- Trinity College Dublin, College Green, Dublin, D2, Ireland
| |
Collapse
|
33
|
Prideaux M, Staines KA, Jones ER, Riley GP, Pitsillides AA, Farquharson C. MMP and TIMP temporal gene expression during osteocytogenesis. Gene Expr Patterns 2015; 18:29-36. [PMID: 25982959 DOI: 10.1016/j.gep.2015.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/20/2015] [Accepted: 04/27/2015] [Indexed: 01/01/2023]
Abstract
Osteocytes within bone differentiate from osteoblast precursors which reside in a mineralised extracellular matrix (ECM). Fully differentiated osteocytes are critical for bone development and function but the factors that regulate this differentiation process are unknown. The enzymes primarily responsible for ECM remodelling are matrix metalloproteinases (MMP); however, the expression and role of MMPs during osteocytogenesis is undefined. Here we used MLO-A5 cells to determine the temporal gene expressions of the MMP family and their endogenous inhibitors--tissue inhibitors of metalloproteinases (TIMPs) during osteocytogenesis. RT-qPCR revealed expression of 14 Mmps and 3 Timps in MLO-A5 cells. Mmp2, Mmp23 and Mmp28 were decreased concurrent with mineralisation onset (P < 0.05*). Mmp14 and Mmp19 mRNAs were also significantly increased at day 3 (P < 0.05*) before returning to baseline levels at day 6. Decreased expressions of Timp1, Timp2 and Timp3 mRNA were observed by day 6 compared to day 0 (P < 0.05*). To examine whether these changes are linked to osteocytogenesis, we determined Mmp/Timp mRNA expressions in mineralisation-limited conditions. RT-qPCR revealed that the previously observed decreases in Mmp2, Mmp23 and Mmp28 were not observed in these mineralisation-limited cultures, therefore closely linking these MMPs with osteocyte differentiation. Similarly, we found differential expression of Timp1, Timp2 and Timp3 mRNA in mineralisation-restricted cultures (P < 0.05*). In conclusion, we have identified several members of the MMP/TIMP families as regulators of ECM remodelling necessary for the acquisition of the osteocyte phenotype.
Collapse
Affiliation(s)
- M Prideaux
- The University of Adelaide, North Terrace, Adelaide, SA 5005, Australia
| | - K A Staines
- Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG.
| | - E R Jones
- University of East Anglia, Norwich NR4 7TJ, UK
| | - G P Riley
- University of East Anglia, Norwich NR4 7TJ, UK
| | - A A Pitsillides
- Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - C Farquharson
- Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG
| |
Collapse
|
34
|
Galea GL, Price JS. Four-point bending protocols to study the effects of dynamic strain in osteoblastic cells in vitro. Methods Mol Biol 2015; 1226:117-130. [PMID: 25331047 DOI: 10.1007/978-1-4939-1619-1_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Strain engendered within bone tissue by mechanical loading of the skeleton is a major influence on the processes of bone modeling and remodeling and so a critical determinant of bone mass and architecture. The cells best placed to respond to strain in bone tissue are the resident osteocytes and osteoblasts. To address the mechanisms of strain-related responses in osteoblast-like cells, our group uses both in vivo and in vitro approaches, including a system of four-point bending of the substrate on which cells are cultured. A range of cell lines can be studied using this system but we routinely compare their responses to those in primary cultures of osteoblast-like cells derived from explants of mouse long bones. These cells show a range of well-characterized responses to physiological levels of strain, including increased proliferation, which in vivo is a feature of the osteogenic response.
Collapse
Affiliation(s)
- Gabriel L Galea
- School of Veterinary Sciences, University of Bristol, Southwell Str, Bristol, BS2 8EJ, UK
| | | |
Collapse
|
35
|
Klein-Nulend J, van Oers RFM, Bakker AD, Bacabac RG. Bone cell mechanosensitivity, estrogen deficiency, and osteoporosis. J Biomech 2014; 48:855-65. [PMID: 25582356 DOI: 10.1016/j.jbiomech.2014.12.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2014] [Indexed: 11/26/2022]
Abstract
Adaptation of bone to mechanical stresses normally produces a bone architecture that combines a proper resistance against failure with a minimal use of material. This adaptive process is governed by mechanosensitive osteocytes that transduce the mechanical signals into chemical responses, i.e. the osteocytes release signaling molecules, which orchestrate the recruitment and activity of bone forming osteoblasts and/or bone resorbing osteoclasts. Computer models have shown that the maintenance of a mechanically-efficient bone architecture depends on the intensity and spatial distribution of the mechanical stimulus as well as on the osteocyte response. Osteoporosis is a condition characterized by a reduced bone mass and a compromized resistance of bone against mechanical loads, which has led us to hypothesize that mechanotransduction by osteocytes is altered in osteoporosis. One of the major causal factors for osteoporosis is the loss of estrogen, the major hormonal regulator of bone metabolism. Loss of estrogen may increase osteocyte-mediated activation of bone remodeling, resulting in impaired bone mass and architecture. In this review we highlight current insights on how osteocytes perceive mechanical stimuli placed on whole bones. Particular emphasis is placed on the role of estrogen in signaling pathway activation by mechanical stimuli, and on computer simulation in combination with cell biology to unravel biological processes contributing to bone strength.
Collapse
Affiliation(s)
- Jenneke Klein-Nulend
- Department of Oral Cell Biology, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands.
| | - René F M van Oers
- Department of Oral Cell Biology, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands; Department of Dental Materials Science, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Astrid D Bakker
- Department of Oral Cell Biology, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Rommel G Bacabac
- Department of Physics, Medical Biophysics Group, University of San Carlos, Cebu City, Philippines
| |
Collapse
|
36
|
Vanderschueren D, Laurent MR, Claessens F, Gielen E, Lagerquist MK, Vandenput L, Börjesson AE, Ohlsson C. Sex steroid actions in male bone. Endocr Rev 2014; 35:906-60. [PMID: 25202834 PMCID: PMC4234776 DOI: 10.1210/er.2014-1024] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sex steroids are chief regulators of gender differences in the skeleton, and male gender is one of the strongest protective factors against osteoporotic fractures. This advantage in bone strength relies mainly on greater cortical bone expansion during pubertal peak bone mass acquisition and superior skeletal maintenance during aging. During both these phases, estrogens acting via estrogen receptor-α in osteoblast lineage cells are crucial for male cortical and trabecular bone, as evident from conditional genetic mouse models, epidemiological studies, rare genetic conditions, genome-wide meta-analyses, and recent interventional trials. Genetic mouse models have also demonstrated a direct role for androgens independent of aromatization on trabecular bone via the androgen receptor in osteoblasts and osteocytes, although the target cell for their key effects on periosteal bone formation remains elusive. Low serum estradiol predicts incident fractures, but the highest risk occurs in men with additionally low T and high SHBG. Still, the possible clinical utility of serum sex steroids for fracture prediction is unknown. It is likely that sex steroid actions on male bone metabolism rely also on extraskeletal mechanisms and cross talk with other signaling pathways. We propose that estrogens influence fracture risk in aging men via direct effects on bone, whereas androgens exert an additional antifracture effect mainly via extraskeletal parameters such as muscle mass and propensity to fall. Given the demographic trends of increased longevity and consequent rise of osteoporosis, an increased understanding of how sex steroids influence male bone health remains a high research priority.
Collapse
Affiliation(s)
- Dirk Vanderschueren
- Clinical and Experimental Endocrinology (D.V.) and Gerontology and Geriatrics (M.R.L., E.G.), Department of Clinical and Experimental Medicine; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine (M.R.L., F.C.); and Centre for Metabolic Bone Diseases (D.V., M.R.L., E.G.), KU Leuven, B-3000 Leuven, Belgium; and Center for Bone and Arthritis Research (M.K.L., L.V., A.E.B., C.O.), Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Wehrle E, Liedert A, Heilmann A, Wehner T, Bindl R, Fischer L, Haffner-Luntzer M, Jakob F, Schinke T, Amling M, Ignatius A. The impact of low-magnitude high-frequency vibration on fracture healing is profoundly influenced by the oestrogen status in mice. Dis Model Mech 2014; 8:93-104. [PMID: 25381012 PMCID: PMC4283653 DOI: 10.1242/dmm.018622] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Fracture healing is impaired in aged and osteoporotic individuals. Because adequate mechanical stimuli are able to increase bone formation, one therapeutical approach to treat poorly healing fractures could be the application of whole-body vibration, including low-magnitude high-frequency vibration (LMHFV). We investigated the effects of LMHFV on fracture healing in aged osteoporotic mice. Female C57BL/6NCrl mice (n=96) were either ovariectomised (OVX) or sham operated (non-OVX) at age 41 weeks. When aged to 49 weeks, all mice received a femur osteotomy that was stabilised using an external fixator. The mice received whole-body vibrations (20 minutes/day) with 0.3 G: peak-to-peak acceleration and a frequency of 45 Hz. After 10 and 21 days, the osteotomised femurs and intact bones (contra-lateral femurs, lumbar spine) were evaluated using bending-testing, micro-computed tomography (μCT), histology and gene expression analyses. LMHFV disturbed fracture healing in aged non-OVX mice, with significantly reduced flexural rigidity (-81%) and bone formation (-80%) in the callus. Gene expression analyses demonstrated increased oestrogen receptor β (ERβ, encoded by Esr2) and Sost expression in the callus of the vibrated animals, but decreased β-catenin, suggesting that ERβ might mediate these negative effects through inhibition of osteoanabolic Wnt/β-catenin signalling. In contrast, in OVX mice, LMHFV significantly improved callus properties, with increased flexural rigidity (+1398%) and bone formation (+637%), which could be abolished by subcutaneous oestrogen application (0.025 mg oestrogen administered in a 90-day-release pellet). On a molecular level, we found an upregulation of ERα in the callus of the vibrated OVX mice, whereas ERβ was unaffected, indicating that ERα might mediate the osteoanabolic response. Our results indicate a major role for oestrogen in the mechanostimulation of fracture healing and imply that LMHFV might only be safe and effective in confined target populations.
Collapse
Affiliation(s)
- Esther Wehrle
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany
| | - Aline Heilmann
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany
| | - Tim Wehner
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany
| | - Ronny Bindl
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany
| | - Lena Fischer
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, 97074 Würzburg, Germany
| | - Thorsten Schinke
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Amling
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, 89081 Ulm, Germany.
| |
Collapse
|
38
|
Kelly NH, Schimenti JC, Patrick Ross F, van der Meulen MCH. A method for isolating high quality RNA from mouse cortical and cancellous bone. Bone 2014; 68:1-5. [PMID: 25073031 PMCID: PMC4281890 DOI: 10.1016/j.bone.2014.07.022] [Citation(s) in RCA: 39] [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] [Received: 06/30/2014] [Revised: 07/18/2014] [Accepted: 07/19/2014] [Indexed: 12/23/2022]
Abstract
The high incidence of fragility fractures in cortico-cancellous bone locations, plus the fact that individual skeletal sites exhibit different responsiveness to load and disease, emphasizes the need to document separately gene expression in cortical and cancellous bone. A further confounding factor is marrow contamination since its high cellularity may effect gene expression measurements. We isolated RNA from cortical and cancellous bone of intact mouse tibiae, and also after marrow removal by flushing or centrifugation. RNA isolated from cancellous bone by each method was sufficient for gene expression analysis. Centrifugation removed contaminating cells more efficiently than flushing, as indexed by histology and decreased expression of Icam4, a highly expressed erythroid gene. In contrast, centrifuged cortical bone had 12- and 13- fold higher expression of the bone-related genes Col1a1 and Bglap, while levels in marrow-free cancellous bone were 30- and 31-fold higher when compared to bone where marrow was left intact. Furthermore, cortical bone had higher expression of Col1a1 and Bglap than cancellous bone. Thus, RNA isolated by this novel approach can reveal site-specific changes in gene expression in cortical and cancellous bone sites.
Collapse
Affiliation(s)
- Natalie H Kelly
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, 105 Upson Hall, Ithaca, NY 14853, USA; Department of Biomedical Engineering, Cornell University, 101 Weill Hall, Ithaca, NY 14853, USA.
| | - John C Schimenti
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - F Patrick Ross
- Research Division, Hospital for Special Surgery, 541 East 71st St., New York, NY 10021, USA.
| | - Marjolein C H van der Meulen
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, 105 Upson Hall, Ithaca, NY 14853, USA; Department of Biomedical Engineering, Cornell University, 101 Weill Hall, Ithaca, NY 14853, USA; Research Division, Hospital for Special Surgery, 541 East 71st St., New York, NY 10021, USA.
| |
Collapse
|
39
|
Morse A, McDonald MM, Kelly NH, Melville KM, Schindeler A, Kramer I, Kneissel M, van der Meulen MCH, Little DG. Mechanical load increases in bone formation via a sclerostin-independent pathway. J Bone Miner Res 2014; 29:2456-67. [PMID: 24821585 PMCID: PMC4501925 DOI: 10.1002/jbmr.2278] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/22/2014] [Accepted: 05/06/2014] [Indexed: 01/24/2023]
Abstract
Sclerostin, encoded by the Sost gene, is an important negative regulator of bone formation that has been proposed to have a key role in regulating the response to mechanical loading. To investigate the effect of long-term Sclerostin deficiency on mechanotransduction in bone, we performed experiments on unloaded or loaded tibiae of 10 week old female Sost-/- and wild type mice. Unloading was induced via 0.5U botulinum toxin (BTX) injections into the right quadriceps and calf muscles, causing muscle paralysis and limb disuse. On a separate group of mice, increased loading was performed on the left tibiae through unilateral cyclic axial compression of equivalent strains (+1200 µe) at 1200 cycles/day, 5 days/week. Another cohort of mice receiving equivalent loads (-9.0 N) also were assessed. Contralateral tibiae served as normal load controls. Loaded/unloaded and normal load tibiae were assessed at day 14 for bone volume (BV) and formation changes. Loss of BV was seen in the unloaded tibiae of wild type mice, but BV was not different between normal load and unloaded Sost-/- tibiae. An increase in BV was seen in the loaded tibiae of wild type and Sost-/- mice over their normal load controls. The increased BV was associated with significantly increased mid-shaft periosteal mineralizing surface/bone surface (MS/BS), mineral apposition rate (MAR), and bone formation rate/bone surface (BFR/BS), and endosteal MAR and BFR/BS. Notably, loading induced a greater increase in periosteal MAR and BFR/BS in Sost-/- mice than in wild type controls. Thus, long-term Sclerostin deficiency inhibits the bone loss normally induced with decreased mechanical load, but it can augment the increase in bone formation with increased load.
Collapse
Affiliation(s)
- A Morse
- Orthopaedic Research and Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
McCarthy TL, Centrella M. Prostaglandin dependent control of an endogenous estrogen receptor agonist by osteoblasts. J Cell Physiol 2014; 230:1104-14. [PMID: 25292157 DOI: 10.1002/jcp.24842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/29/2014] [Indexed: 12/30/2022]
Abstract
Estrogen receptor (ER) activation has complex effects on bone cells, and loss of circulating estradiol adversely affects skeletal status in women. Hormone replacement therapy effectively circumvents bone loss after menopause, but enhances disease risk in other tissues. Here we show that prostaglandin E2 (PGE2) augments the activity of an osteoblast-derived selective ER modulator, ObSERM. The stimulatory effect of PGE2 is replicated in part by either the PG receptor EP3 agonist 17-phenyl trinor PGE2 or by the PG receptor FP agonist PGF2α⋅ Whereas activation of the various PG receptors induces multiple downstream signals, the response to PGE2 was mimicked by activators of protein kinase C, and suppressed by inhibition of protein kinase C but not by inhibition of protein kinase A. Moreover, inhibition of nitric oxide synthesis and activation of the PTH and Wnt pathways increases ObSERM activity. Our studies therefore reveal that ObSERM activity is controlled in distinct ways and revise our understanding of ER activation within bone by agents or events associated with PG expression. They also predict ways to sustain or improve bone formation, fracture repair, and surgical healing without adding the risk of disease in other tissues where ER activation also has important biological functions.
Collapse
Affiliation(s)
- Thomas L McCarthy
- Departments of Pathology and Surgery, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
41
|
Meakin LB, Galea GL, Sugiyama T, Lanyon LE, Price JS. Age-related impairment of bones' adaptive response to loading in mice is associated with sex-related deficiencies in osteoblasts but no change in osteocytes. J Bone Miner Res 2014; 29:1859-71. [PMID: 24644060 PMCID: PMC4258100 DOI: 10.1002/jbmr.2222] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/23/2014] [Accepted: 03/03/2014] [Indexed: 01/08/2023]
Abstract
Bones adjust their mass and architecture to be sufficiently robust to withstand functional loading by adapting to their strain environment. This mechanism appears less effective with age, resulting in low bone mass. In male and female young adult (17-week-old) and old (19-month-old) mice, we investigated the effect of age in vivo on bones' adaptive response to loading and in vitro in primary cultures of osteoblast-like cells derived from bone cortices. Right tibias were axially loaded on alternate days for 2 weeks. Left tibias were non-loaded controls. In a separate group, the number of sclerostin-positive osteocytes and the number of periosteal osteoblasts were analyzed 24 hours after a single loading episode. The responses to strain of the primary osteoblast-like cells derived from these mice were assessed by EGR2 expression, change in cell number and Ki67 immunofluorescence. In young male and female mice, loading increased trabecular thickness and the number of trabecular connections. Increase in the number of trabecular connections was impaired with age but trabecular thickness was not. In old mice, the loading-related increase in periosteal apposition of the cortex was less than in young ones. Age was associated with a lesser loading-related increase in osteoblast number on the periosteal surface but had no effect on loading-related reduction in the number of sclerostin-positive osteocytes. In vitro, strain-related proliferation of osteoblast-like cells was lower in cells from old than young mice. Cells from aged female mice demonstrated normal entry into the cell cycle but subsequently arrested in G2 phase, reducing strain-related increases in cell number. Thus, in both male and female mice, loading-related adaptive responses are impaired with age. This impairment is different in females and males. The deficit appears to occur in osteoblasts' proliferative responses to strain rather than earlier strain-related responses in the osteocytes.
Collapse
Affiliation(s)
- Lee B Meakin
- School of Veterinary Science, University of BristolLangford, Bristol, UK
| | - Gabriel L Galea
- School of Veterinary Science, University of BristolLangford, Bristol, UK
| | - Toshihiro Sugiyama
- School of Veterinary Science, University of BristolLangford, Bristol, UK
| | - Lance E Lanyon
- School of Veterinary Science, University of BristolLangford, Bristol, UK
| | - Joanna S Price
- School of Veterinary Science, University of BristolLangford, Bristol, UK
| |
Collapse
|
42
|
Klein-Nulend J, van Oers RFM, Bakker AD, Bacabac RG. Nitric oxide signaling in mechanical adaptation of bone. Osteoporos Int 2014; 25:1427-37. [PMID: 24322479 DOI: 10.1007/s00198-013-2590-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/24/2013] [Indexed: 01/27/2023]
Abstract
One of the most serious healthcare problems in the world is bone loss and fractures due to a lack of physical activity in elderly people as well as in bedridden patients or otherwise inactive youth. Crucial here are the osteocytes. Buried within our bones, these cells are believed to be the mechanosensors that stimulate bone formation in the presence of mechanical stimuli and bone resorption in the absence of such stimuli. Intercellular signaling is an important physiological phenomenon involved in maintaining homeostasis in all tissues. In bone, intercellular communication via chemical signals like NO plays a critical role in the dynamic process of bone remodeling. If bones are mechanically loaded, fluid flows through minute channels in the bone matrix, resulting in shear stress on the cell membrane that activates the osteocyte. Activated osteocytes produce signaling molecules like NO, which modulate the activity of the bone-forming osteoblasts and the bone-resorbing osteoclasts, thereby orchestrating bone adaptation to mechanical loading. In this review, we highlight current insights in the role of NO in the mechanical adaptation of bone mass and structure, with emphasis on its role in local bone gain and loss as well as in remodeling supervised by osteocytes. Since mechanical stimuli and NO production enhance bone strength and fracture resistance, these new insights may facilitate the development of novel osteoporosis treatments.
Collapse
Affiliation(s)
- J Klein-Nulend
- Department of Oral Cell Biology, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Gustav Mahlerlaan 3004, 1081, LA, Amsterdam, The Netherlands,
| | | | | | | |
Collapse
|
43
|
Ciani C, Sharma D, Doty SB, Fritton SP. Ovariectomy enhances mechanical load-induced solute transport around osteocytes in rat cancellous bone. Bone 2014; 59:229-34. [PMID: 24316418 PMCID: PMC4358819 DOI: 10.1016/j.bone.2013.11.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 11/08/2013] [Accepted: 11/26/2013] [Indexed: 01/06/2023]
Abstract
To test if osteoporosis alters mechanical load-induced interstitial fluid flow in bone, this study examined the combined effect of estrogen deficiency and external loading on solute transport around osteocytes. An in vivo tracer, FITC-labeled bovine serum albumin, was injected into anesthetized ovariectomized and control female Sprague-Dawley rats before the right tibia was subjected to a controlled, physiological, non-invasive sinusoidal load to mimic walking. Tracer movement through the lacunar-canalicular system surrounding osteocytes was quantified in cortical and cancellous bone from the proximal tibia using confocal microscopy, with the non-loaded tibia serving as internal control. Overall, the application of mechanical loading increased the percentage of osteocyte lacunae labeled with injected tracer, and ovariectomy further enhanced movement of tracer. An analysis of separate regions demonstrated that ovariectomy enhanced in vivo transport of the injected tracer in the cancellous bone of the tibial epiphysis and metaphysis but not in the cortical bone of the metaphysis. These findings show that bone changes due to reduced estrogen levels alter convectional transport around osteocytes in cancellous bone and demonstrate a functional difference of interstitial fluid flow around osteocytes in estrogen-deficient rats undergoing the same physical activity as controls. The altered interstitial fluid flow around osteocytes is likely related to nanostructural matrix-mineral level differences recently demonstrated at the lacunar-canalicular surface of estrogen-deficient rats, which could affect the transmission of mechanical loads to the osteocyte.
Collapse
Affiliation(s)
- Cesare Ciani
- Department of Biomedical Engineering, City College of New York, New York, NY 10031, USA
| | - Divya Sharma
- Department of Biomedical Engineering, City College of New York, New York, NY 10031, USA
| | - Stephen B Doty
- Research Division, Hospital for Special Surgery, New York, NY 10021, USA
| | - Susannah P Fritton
- Department of Biomedical Engineering, City College of New York, New York, NY 10031, USA.
| |
Collapse
|
44
|
Meakin LB, Price JS, Lanyon LE. The Contribution of Experimental in vivo Models to Understanding the Mechanisms of Adaptation to Mechanical Loading in Bone. Front Endocrinol (Lausanne) 2014; 5:154. [PMID: 25324829 PMCID: PMC4181237 DOI: 10.3389/fendo.2014.00154] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/12/2014] [Indexed: 12/12/2022] Open
Abstract
Changing loading regimens by natural means such as exercise, with or without interference such as osteotomy, has provided useful information on the structure:function relationship in bone tissue. However, the greatest precision in defining those aspects of the overall strain environment that influence modeling and remodeling behavior has been achieved by relating quantified changes in bone architecture to quantified changes in bones' strain environment produced by direct, controlled artificial bone loading. Jiri Hert introduced the technique of artificial loading of bones in vivo with external devices in the 1960s using an electromechanical device to load rabbit tibiae through transfixing stainless steel pins. Quantifying natural bone strains during locomotion by attaching electrical resistance strain gages to bone surfaces was introduced by Lanyon, also in the 1960s. These studies in a variety of bones in a number of species demonstrated remarkable uniformity in the peak strains and maximum strain rates experienced. Experiments combining strain gage instrumentation with artificial loading in sheep, pigs, roosters, turkeys, rats, and mice has yielded significant insight into the control of strain-related adaptive (re)modeling. This diversity of approach has been largely superseded by non-invasive transcutaneous loading in rats and mice, which is now the model of choice for many studies. Together such studies have demonstrated that over the physiological strain range, bone's mechanically adaptive processes are responsive to dynamic but not static strains; the size and nature of the adaptive response controlling bone mass is linearly related to the peak loads encountered; the strain-related response is preferentially sensitive to high strain rates and unresponsive to static ones; is most responsive to unusual strain distributions; is maximized by remarkably few strain cycles, and that these are most effective when interrupted by short periods of rest between them.
Collapse
Affiliation(s)
- Lee B. Meakin
- School of Veterinary Sciences, University of Bristol, Bristol, UK
- *Correspondence: Lee B. Meakin, School of Veterinary Sciences, University of Bristol, Langford House, Langford, Bristol BS40 5DU, UK e-mail:
| | - Joanna S. Price
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Lance E. Lanyon
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
45
|
Shwartz Y, Blitz E, Zelzer E. One load to rule them all: Mechanical control of the musculoskeletal system in development and aging. Differentiation 2013; 86:104-11. [DOI: 10.1016/j.diff.2013.07.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 07/01/2013] [Accepted: 07/12/2013] [Indexed: 12/24/2022]
|
46
|
Judex S, Zhang W, Donahue LR, Ozcivici E. Genetic loci that control the loss and regain of trabecular bone during unloading and reambulation. J Bone Miner Res 2013; 28:1537-49. [PMID: 23401066 DOI: 10.1002/jbmr.1883] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/15/2013] [Accepted: 01/23/2013] [Indexed: 12/18/2022]
Abstract
Changes in trabecular morphology during unloading and reloading are marked by large variations between individuals, implying that there is a strong genetic influence on the magnitude of the response. Here, we subjected more than 350 second-generation (BALBxC3H) 4-month-old adult female mice to 3 weeks of hindlimb unloading followed by 3 weeks of reambulation to identify the quantitative trait loci (QTLs) that define an individual's propensity to either lose trabecular bone when weight bearing is removed or to gain trabecular bone when weight bearing is reintroduced. Longitudinal in vivo micro-computed tomography (µCT) scans demonstrated that individual mice lost between 15% and 71% in trabecular bone volume fraction (BV/TV) in the distal femur during unloading (average: -43%). Changes in trabecular BV/TV during the 3-week reambulation period ranged from a continuation of bone loss (-18%) to large additions (56%) of tissue (average: +10%). During unloading, six QTLs accounted for 21% of the total variability in changes in BV/TV whereas one QTL accounted for 6% of the variability in changes in BV/TV during reambulation. QTLs were also identified for changes in trabecular architecture. Most of the QTLs defining morphologic changes during unloading or reambulation did not overlap with those QTLs identified at baseline, suggesting that these QTLs harbor genes that are specific for sensing changes in the levels of weight bearing. The lack of overlap in QTLs between unloading and reambulation also emphasizes that the genes modulating the trabecular response to unloading are distinct from those regulating tissue recovery during reloading. The identified QTLs contain the regulatory genes underlying the strong genetic regulation of trabecular bone's sensitivity to weight bearing and may help to identify individuals that are most susceptible to unloading-induced bone loss and/or the least capable of recovering.
Collapse
Affiliation(s)
- Stefan Judex
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | | | | | | |
Collapse
|
47
|
Wu X, Guo S, Shen G, Ma X, Tang C, Xie K, Liu J, Guo W, Yan Y, Luo E. Screening of osteoprotegerin-related feature genes in osteoporosis and functional analysis with DNA microarray. Eur J Med Res 2013; 18:15. [PMID: 23731710 PMCID: PMC3735399 DOI: 10.1186/2047-783x-18-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 05/06/2013] [Indexed: 11/26/2022] Open
Abstract
Background Osteoporosis affects 200 million people worldwide and places an enormous economic burden on society. We aim to identify the feature genes that are related to osteoprotegerin in osteoporosis and to perform function analysis with DNA microarray from human bone marrow. Methods We downloaded the gene expression profile GSE35957 from Gene Expression Omnibus database including nine gene chips from bone marrow mesenchymal stem cells of five osteoporotic and four non-osteoporotic subjects. The differentially expressed genes between normal and disease samples were identified by LIMMA package in R language. The interactions among the osteoprotegerin gene (OPG) and differentially expressed genes were searched and visualized by Cytoscape. MCODE and Bingo were used to perform module analysis. Finally, GENECODIS was used to obtain enriched pathways of genes in an interaction network. Results A total of 656 genes were identified as differentially expressed genes between osteoporotic and non-osteoporotic samples. IL17RC, COL1A1, and ESR1 were identified to interact with OPG directly from the protein-protein interaction network. A module containing ERS1 was screened out, and this module was most significantly enriched in organ development. Pathway enrichment analysis suggested genes in the interaction network were related to focal adhesion. Conclusions The expression pattern of IL17RC, COL1A1, and ESR1 can be useful in osteoporosis detection, which may help in identifying those populations at high risk for osteoporosis, and in directing treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiaoming Wu
- School of Biomedical Engineering, Fourth Military Medical University, No, 17, Changle West Road, Xi'an, Shanxi 710032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sapir-Koren R, Livshits G. Is interaction between age-dependent decline in mechanical stimulation and osteocyte-estrogen receptor levels the culprit for postmenopausal-impaired bone formation? Osteoporos Int 2013; 24:1771-89. [PMID: 23229466 DOI: 10.1007/s00198-012-2208-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 10/02/2012] [Indexed: 12/19/2022]
Abstract
Declining estrogen levels during menopause are widely considered to be a major cause of age-dependent bone loss, which is primarily manifested by increased bone resorption by osteoclasts. We present accumulating evidence supporting another aspect of metabolic bone loss, suggesting that the combined interaction between age-dependent factors, namely, estrogen deficiency and reduced day-by-day activity/mechanical stimulation, directly leads to a reduction in anabolic processes. Such decreased bone formation results in diminished bone strength and failure to maintain the load-bearing competence of a healthy skeleton and to postmenopausal osteoporosis disorder. Estrogen receptors (ERs), as mediators of estrogenic actions, are essential components of bone osteocyte and osteoblast mechano-adaptive responses. ER expression appears to be upregulated by adequate circulating estrogen levels. ERα signaling pathways participate in the mechanotransduction response through obligatory "non-genomic" actions that occur independently of estrogen binding to ER and by a potentially "genomic", estrogen-dependent mode. The experimental data indicate that cross talk between the ERα-"non-genomic" and Wnt/β-catenin signaling pathways constitutes the major regulatory mechanism. This interaction uses mechanically and ER-induced prostaglandin E2 as a mediator for the downregulation of osteocyte production of sclerostin. Sclerostin suppression, in turn, is a central prerequisite for load-induced formation and mineralization of the bone matrix. It is therefore plausible that future strategies for preventing and treating postmenopausal osteoporosis may use estrogenic compounds (such as selective estrogen receptor modulators or phytoestrogens) with physical activity, to complement antiresorptive therapy, aimed at stopping further bone loss and possibly even reversing it by stimulation of bone gain.
Collapse
Affiliation(s)
- R Sapir-Koren
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | | |
Collapse
|
49
|
Tsuchiya T, Sakai A, Menuki K, Mori T, Takeuchi Y, Kanoh S, Utsunomiya H, Murai T, Isse T, Kawamoto T, Nakamura T. Disruption of aldehyde dehydrogenase 2 gene results in altered cortical bone structure and increased cortical bone mineral density in the femoral diaphysis of mice. Bone 2013; 53:358-68. [PMID: 23313283 DOI: 10.1016/j.bone.2012.12.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/18/2012] [Accepted: 12/28/2012] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Aldehyde dehydrogenase 2 (ALDH2) degrades acetaldehyde produced by the metabolism of alcohol. The inactive ALDH2 phenotype is prevalent in East Asians, and an association between this ALDH2 polymorphism and osteoporosis has been reported. In our previous study, we found that alcohol consumption results in decreased trabecular bone volume in aldh2 knockout (aldh2(-/-)) mice compared with the volume in wild-type (aldh2(+/+)) mice. However, the effect of aldh2 gene on the skeletal phenotype in the absence of alcohol consumption remains unknown. The aim of this study was to clarify the effect of aldh2 disruption on femoral bone structure and dynamics in aldh2-disrupted mice in the absence of alcohol consumption. MATERIALS AND METHODS We examined aldh2(-/-) and aldh2(+/+) mice at the ages of 4, 8 and 12weeks. The femoral bone length and bone mineral density (BMD) were measured using peripheral quantitative computed tomography. The mechanical strength was assessed by the three-point bending test at 12weeks, and cortical bone histomorphometry at the femur diaphysis was performed at all three time points. Osteogenic activities in aldh2(-/-) and aldh2(+/+) mice were assessed by osteoblast culture from calvariae of the neonatal mice. Bilateral femoral and tibial bones containing no bone marrow cells of 8-week-old mice were used for analysis of mRNA expression. In addition, mRNA expression in aldh2(-/-) and aldh2(+/+) mice after tail suspension or climbing exercise for 7days from 8weeks was analyzed to clarify the response to mechanical loading. RESULTS At 12weeks, there were no significant differences in femoral bone length, trabecular BMD in the distal metaphyses of the femurs, or mechanical strength between aldh2(-/-) and aldh2(+/)(+) mice, whereas cortical BMD and cortical thickness were significantly increased and cross-sectional area and bone marrow area were significantly decreased in the femoral diaphysis of aldh2(-/-) mice relative to the corresponding values in aldh2(+/+) mice. At 8weeks, bone formation rate and mineral apposition rate on the periosteal and endocortical surfaces were significantly increased in aldh2(-/-) mice relative to the rates in aldh(+/+) mice. Calvarial osteoblast culture study revealed that the percentage of alkaline phosphatase stained cells was significantly higher in aldh2(-/-) mice compared to that in aldh(+/+) mice. Quantitative real-time RT-PCR revealed a significant increase in the expressions of bmp2, osterix, runx2, and col1a1 mRNA in aldh2(-/-) mice, along with an increase in the expression of wnt5a mRNA and the lrp5/sost mRNA ratio. The mRNA expressions of bmp2, osterix, runx2 and pthr in aldh2(-/-) mice were significantly decreased after climbing exercise compared to those in the control, although the mRNA expressions of bmp2, osterix, runx2 were not significantly decreased and pthr mRNA expression was increased in aldh(+/+) mice after climbing exercise. CONCLUSION Our results show that disruption of aldh2 gene resulted in altered cortical bone structure and dynamics in mice. Cross-sectional area was decreased. Cortical BMD was increased owing to the promotion of cortical bone formation on the periosteal and endocortical surfaces of the femoral diaphysis. The possible mechanisms underlying altered cortical bone structure in aldh2(-/-) mice were gene-related higher osteogenic activity of osteoblasts and weakened osteogenice response to mechanical loading in growth period.
Collapse
Affiliation(s)
- Takuto Tsuchiya
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Macias BR, Aspenberg P, Agholme F. Paradoxical Sost gene expression response to mechanical unloading in metaphyseal bone. Bone 2013; 53:515-9. [PMID: 23337040 DOI: 10.1016/j.bone.2013.01.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/10/2013] [Accepted: 01/11/2013] [Indexed: 01/20/2023]
Abstract
The Sost gene encodes Sclerostin, an inhibitor of Wnt-signaling, generally considered a main response gene to mechanical loading in bone. Several papers describe that unloading leads to upregulation of Sost, which in turn may lead to loss of bone. These studies were based on whole bone homogenates or cortical bone. By serendipity, we noted an opposite response to unloading in the proximal rat tibia. Therefore, we hypothesized that Sost-expression in response to changes in mechanical load is bone site specific. One hind limb of male, 3 month old rats was unloaded by paralyzing the extensors with Botulinium toxin A (Botox) injections. A series of experiments compared the expression of Sost mRNA in the unloaded and contralateral, loaded limbs, after 3 or 10 days, in metaphyseal cancellous bone, metaphyseal cortical bone, and diaphyseal cortical bone. We also conducted μCT to confirm changes in bone volume density related to unloading. Sost mRNA expression in the cancellous metaphyseal bone was downregulated almost 2-fold, both 3 days and 10 days after unloading (P<0.05). A similar tendency was seen in the metaphyseal cortical bone, in which Sost was 1.5-fold downregulated (P<0.05) after 10days, but not significantly changed after 3days. In contrast, diaphyseal cortical Sost expression was instead upregulated 1.4-fold (P<0.05) following 3-day unloading, while there was no significant change after 10days. Cancellous bone volume density was 58% lower (P<0.001, compared to cage controls) in the unloaded limb but not significantly affected in the loaded limb. The results suggest that Sost mRNA expression in metaphyseal bone responds to mechanical unloading in an opposite direction to that observed in diaphyseal cortical bone. This proposes a more complex expression pattern for Sost in response to unloading. Therapeutics that target Sclerostin during altered loading conditions may result in local bone mass changes that are difficult to predict.
Collapse
Affiliation(s)
- Brandon R Macias
- Orthopedics, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden.
| | | | | |
Collapse
|