1
|
Valverde A, George A, Nares S, Naqvi AR. Emerging therapeutic strategies targeting bone signaling pathways in periodontitis. J Periodontal Res 2025; 60:101-120. [PMID: 39044454 PMCID: PMC11873684 DOI: 10.1111/jre.13326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/22/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024]
Abstract
Periodontitis is a multifactorial immune-mediated disease exacerbated by dysregulated alveolar bone homeostasis. Timely intervention is crucial for disease management to prevent tooth loss. To successfully manage periodontitis, it is imperative to understand the cellular and molecular mechanisms involved in its pathogenesis to develop novel treatment modalities. Non-surgical periodontal therapy (NSPT) such as subgingival instrumentation/debridement has been the underlying treatment strategy over the past decades. However, new NSPT approaches that target key signaling pathways regulating alveolar bone homeostasis have shown positive clinical outcomes. This narrative review aims to discuss endogenous bone homeostasis mechanisms impaired in periodontitis and highlight the clinical outcomes of preventive periodontal therapy to avoid invasive periodontal therapies. Although the anti-resorptive therapeutic adjuncts have demonstrated beneficial outcomes, adverse events have been reported. Diverse immunomodulatory therapies targeting the osteoblast/osteoclast (OB/OC) axis have shown promising outcomes in vivo. Future controlled randomized clinical trials (RCT) would help clinicians and patients in the selection of novel preventing therapies targeting key molecules to effectively treat or prevent periodontitis.
Collapse
Affiliation(s)
- Araceli Valverde
- Department of PeriodonticsCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
| | - Anne George
- Department of Oral BiologyCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
| | - Salvador Nares
- Department of PeriodonticsCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
| | - Afsar R. Naqvi
- Department of PeriodonticsCollege of Dentistry, University of Illinois ChicagoChicagoIllinoisUSA
- Department of Microbiology and ImmunologyUniversity of Illinois ChicagoChicagoIllinoisUSA
| |
Collapse
|
2
|
Lin SC, Yu G, Corn PG, Damasco J, Lee YC, Song JH, Navone NM, Logothetis CJ, Melancon MP, Panaretakis T, Lin SH. Radium-223 Treatment Produces Prolonged Suppression of Resident Osteoblasts and Decreased Bone Mineral Density in Trabecular Bone in Osteoblast Reporter Mice. Cancers (Basel) 2024; 16:2603. [PMID: 39061241 PMCID: PMC11274981 DOI: 10.3390/cancers16142603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/21/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Radium 223 (Ra-223) is an α-emitting bone-homing radiopharmaceutical that targets tumor-induced osteoblasts and is used to reduce bone pain and prolong overall survival in men with bone-metastatic, castrate-resistant prostate cancer. However, increased fracture risk in skeletal sites with no bone metastasis has been observed in patients treated with Ra-223. Both luciferase- or green fluorescence protein (GFP)-labeled osteoblast reporter mice were used to monitor the effect of Ra-223 on resident osteoblasts and normal bone structure. Upon Ra-223 treatment, 70% of resident osteoblasts were reduced within 2 days, and the osteoblast reduction lasted for at least 18 weeks without detectable recovery, as measured by in vivo bioluminescent imaging. In GFP-labeled osteoblast reporter mice, Ra-223 mainly reduced osteoblasts localized in the trabecular bone areas; the osteoblasts in the growth plates were less affected. Micro-computed tomography analyses showed that Ra-223 significantly reduced bone mineral density and bone microstructure in the trabecular area of femurs but not in the cortical bone. Tumor-induced bone was generated by inoculating osteogenic TRAMP-BMP4 prostate cancer cells into the mouse femurs; Ra-223 treatment significantly reduced tumor-induced osteoblasts. Our study shows that Ra-223 affects bone structures that are not involved in bone metastasis. Strategies that improve bone health may reduce fracture risk in patients receiving Ra-223.
Collapse
Affiliation(s)
- Song-Chang Lin
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (S.-C.L.); (G.Y.); (Y.-C.L.)
| | - Guoyu Yu
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (S.-C.L.); (G.Y.); (Y.-C.L.)
| | - Paul G. Corn
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (P.G.C.); (J.H.S.); (N.M.N.); (C.J.L.)
| | - Jossana Damasco
- Department of Interventional Radiology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (J.D.); (M.P.M.)
| | - Yu-Chen Lee
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (S.-C.L.); (G.Y.); (Y.-C.L.)
| | - Jian H. Song
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (P.G.C.); (J.H.S.); (N.M.N.); (C.J.L.)
| | - Nora M. Navone
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (P.G.C.); (J.H.S.); (N.M.N.); (C.J.L.)
| | - Christopher J. Logothetis
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (P.G.C.); (J.H.S.); (N.M.N.); (C.J.L.)
| | - Marites P. Melancon
- Department of Interventional Radiology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (J.D.); (M.P.M.)
- UTHealth Houston Graduate School of Biomedical Sciences, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (P.G.C.); (J.H.S.); (N.M.N.); (C.J.L.)
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (S.-C.L.); (G.Y.); (Y.-C.L.)
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA; (P.G.C.); (J.H.S.); (N.M.N.); (C.J.L.)
- UTHealth Houston Graduate School of Biomedical Sciences, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| |
Collapse
|
3
|
Reddien PW. The purpose and ubiquity of turnover. Cell 2024; 187:2657-2681. [PMID: 38788689 DOI: 10.1016/j.cell.2024.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
Turnover-constant component production and destruction-is ubiquitous in biology. Turnover occurs across organisms and scales, including for RNAs, proteins, membranes, macromolecular structures, organelles, cells, hair, feathers, nails, antlers, and teeth. For many systems, turnover might seem wasteful when degraded components are often fully functional. Some components turn over with shockingly high rates and others do not turn over at all, further making this process enigmatic. However, turnover can address fundamental problems by yielding powerful properties, including regeneration, rapid repair onset, clearance of unpredictable damage and errors, maintenance of low constitutive levels of disrepair, prevention of stable hazards, and transitions. I argue that trade-offs between turnover benefits and metabolic costs, combined with constraints on turnover, determine its presence and rates across distinct contexts. I suggest that the limits of turnover help explain aging and that turnover properties and the basis for its levels underlie this fundamental component of life.
Collapse
Affiliation(s)
- Peter W Reddien
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
4
|
Guo W, Hoque J, Garcia CJG, Spiller KV, Leinroth AP, Puviindran V, Potnis CK, Gunn KA, Newman H, Ishikawa K, Fujimoto TN, Neill DW, Delahoussaye AM, Williams NT, Kirsch DG, Hilton MJ, Varghese S, Taniguchi CM, Wu C. Radiation-induced bone loss in mice is ameliorated by inhibition of HIF-2α in skeletal progenitor cells. Sci Transl Med 2023; 15:eabo5217. [PMID: 38019933 PMCID: PMC10804914 DOI: 10.1126/scitranslmed.abo5217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 10/16/2023] [Indexed: 12/01/2023]
Abstract
Radiotherapy remains a common treatment modality for cancer despite skeletal complications. However, there are currently no effective treatments for radiation-induced bone loss, and the consequences of radiotherapy on skeletal progenitor cell (SPC) survival and function remain unclear. After radiation, leptin receptor-expressing cells, which include a population of SPCs, become localized to hypoxic regions of the bone and stabilize the transcription factor hypoxia-inducible factor-2α (HIF-2α), thus suggesting a role for HIF-2α in the skeletal response to radiation. Here, we conditionally knocked out HIF-2α in leptin receptor-expressing cells and their descendants in mice. Radiation therapy in littermate control mice reduced bone mass; however, HIF-2α conditional knockout mice maintained bone mass comparable to nonirradiated control animals. HIF-2α negatively regulated the number of SPCs, bone formation, and bone mineralization. To test whether blocking HIF-2α pharmacologically could reduce bone loss during radiation, we administered a selective HIF-2α inhibitor called PT2399 (a structural analog of which was recently FDA-approved) to wild-type mice before radiation exposure. Pharmacological inhibition of HIF-2α was sufficient to prevent radiation-induced bone loss in a single-limb irradiation mouse model. Given that ~90% of patients who receive a HIF-2α inhibitor develop anemia because of off-target effects, we developed a bone-targeting nanocarrier formulation to deliver the HIF-2α inhibitor to mouse bone, to increase on-target efficacy and reduce off-target toxicities. Nanocarrier-loaded PT2399 prevented radiation-induced bone loss in mice while reducing drug accumulation in the kidney. Targeted inhibition of HIF-2α may represent a therapeutic approach for protecting bone during radiation therapy.
Collapse
Affiliation(s)
- Wendi Guo
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine; Durham, NC 27705, USA
| | - Jiaul Hoque
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
| | - Carolina J. Garcia Garcia
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center; Houston, TX 77030, USA
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX 77030, USA
| | - Kassandra V. Spiller
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27705, USA
| | - Abigail P. Leinroth
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | - Vijitha Puviindran
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
| | - Cahil K. Potnis
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
| | - Kiana A. Gunn
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine; Durham, NC 27705, USA
| | - Hunter Newman
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Mechanical Engineering and Materials Science, Duke University; Durham, NC 27705, USA
| | - Koji Ishikawa
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Orthopaedic Surgery, Showa University School of Medicine, Tokyo, 142-8666, JP
| | - Tara N. Fujimoto
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX 77030, USA
| | - Denae W. Neill
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center; Houston, TX 77030, USA
| | - Abagail M. Delahoussaye
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX 77030, USA
| | - Nerissa T. Williams
- Department of Radiation Oncology, Duke University School of Medicine; Durham, NC 27705, USA
| | - David G. Kirsch
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Radiation Oncology, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Biomedical Biophysics, University of Toronto, Toronto, ON, M5S 1A8, CA
- Department of Radiation Oncology, University of Toronto, Toronto, ON, M5T 1O5, CA
- Princess Margarat Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, CA
| | - Matthew J. Hilton
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Cell Biology, Duke University School of Medicine; Durham, NC 27705, USA
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Mechanical Engineering and Materials Science, Duke University; Durham, NC 27705, USA
- Department of Biomedical Engineering, Duke University School of Medicine; Durham, NC 27705, USA
| | - Cullen M. Taniguchi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX 77030, USA
- Department of GI Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX 77030, USA
| | - Colleen Wu
- Department of Orthopaedic Surgery, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine; Durham, NC 27705, USA
- Department of Radiation Oncology, University of Toronto, Toronto, ON, M5T 1O5, CA
| |
Collapse
|
5
|
Ordaz G, Rentería JA, Mariscal G. Characterization and modeling of the serum concentration of osteocalcin in breeding sows and its interaction with biochemical indicators: A review. J Adv Vet Anim Res 2022; 9:634-648. [PMID: 36714515 PMCID: PMC9868799 DOI: 10.5455/javar.2022.i633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 01/13/2023] Open
Abstract
Adipose, muscle, and bone tissues modulate the metabolic state of mammals. However, the role of bone tissue as a metabolic state modulator in sows has not been studied. During the gestation-lactation transition, sows undergo metabolic adaptations to meet their nutritional requirements. Among these adaptations, bone remodeling is characterized by the synthesis and inhibition of hormones that participate, together with hormones from other tissues, in fetal development and lactogenesis. Osteocalcin is a hormone synthesized by the bone tissue which has been associated in different biological models with the improvement of the metabolic state. However, in sows, published results on the concentration of osteocalcin are scarce, and its concentration throughout the reproductive cycle is unknown. Therefore, with information from published trials on the measurement of serum osteocalcin, a structured review was conducted under the following objectives: (1) to review the promising effect of osteocalcin on energy metabolism in different models and (2) to characterize and model the serum concentrations of osteocalcin during the reproductive cycle of the sow. According to the review, the results obtained for humans and other animal models suggest that osteocalcin regulates energy metabolism, which has been associated with the need for integrated metabolism to cope with the metabolic demand during gestation and lactation in mammals. If these effects are significant in the sow, current recommendations for dietary balance should be reconsidered, particularly during the gestation-lactation transition period. According to mathematical modeling, it was the period in which the lowest concentration of osteocalcin was found.
Collapse
Affiliation(s)
- Gerardo Ordaz
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal-INIFAP, Querétaro, México
| | - José Antonio Rentería
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal-INIFAP, Querétaro, México
| | - Gerardo Mariscal
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal-INIFAP, Querétaro, México
| |
Collapse
|
6
|
Kim MJ, Valderrábano RJ, Wu JY. Osteoblast Lineage Support of Hematopoiesis in Health and Disease. J Bone Miner Res 2022; 37:1823-1842. [PMID: 35983701 PMCID: PMC11346465 DOI: 10.1002/jbmr.4678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/21/2022] [Accepted: 08/13/2022] [Indexed: 11/06/2022]
Abstract
In mammals, hematopoiesis migrates to the bone marrow during embryogenesis coincident with the appearance of mineralized bone, where hematopoietic stem cells (HSCs) and their progeny are maintained by the surrounding microenvironment or niche, and sustain the entirety of the hematopoietic system. Genetic manipulation of niche factors and advances in cell lineage tracing techniques have implicated cells of both hematopoietic and nonhematopoietic origin as important regulators of hematopoiesis in health and disease. Among them, cells of the osteoblast lineage, from stromal skeletal stem cells to matrix-embedded osteocytes, are vital niche residents with varying capacities for hematopoietic support depending on stage of differentiation. Here, we review populations of osteoblasts at differing stages of differentiation and summarize the current understanding of the role of the osteoblast lineage in supporting hematopoiesis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Matthew J Kim
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rodrigo J Valderrábano
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joy Y Wu
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
7
|
Gultian KA, Gandhi R, DeCesari K, Romiyo V, Kleinbart EP, Martin K, Gentile PM, Kim TWB, Vega SL. Injectable hydrogel with immobilized BMP-2 mimetic peptide for local bone regeneration. FRONTIERS IN BIOMATERIALS SCIENCE 2022; 1. [PMID: 37090104 PMCID: PMC10120851 DOI: 10.3389/fbiom.2022.948493] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Osteoporosis is a disease characterized by a decrease in bone mineral density, thereby increasing the risk of sustaining a fragility fracture. Most medical therapies are systemic and do not restore bone in areas of need, leading to undesirable side effects. Injectable hydrogels can locally deliver therapeutics with spatial precision, and this study reports the development of an injectable hydrogel containing a peptide mimic of bone morphogenetic protein-2 (BMP-2). To create injectable hydrogels, hyaluronic acid was modified with norbornene (HANor) or tetrazine (HATet) which upon mixing click into covalently crosslinked Nor-Tet hydrogels. By modifying HANor macromers with methacrylates (Me), thiolated BMP-2 mimetic peptides were immobilized to HANor via a Michael addition reaction, and coupling was confirmed with 1H NMR spectroscopy. BMP-2 peptides presented in soluble and immobilized form increased alkaline phosphatase (ALP) expression in MSCs cultured on 2D and encapsulated in 3D Nor-Tet hydrogels. Injection of bioactive Nor-Tet hydrogels into hollow intramedullary canals of Lewis rat femurs showed a local increase in trabecular bone density as determined by micro-CT imaging. The presented work shows that injectable hydrogels with immobilized BMP-2 peptides are a promising biomaterial for the local regeneration of bone tissue and for the potential local treatment of osteoporosis.
Collapse
Affiliation(s)
- Kirstene A. Gultian
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
| | - Roshni Gandhi
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
| | - Kayla DeCesari
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
| | - Vineeth Romiyo
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Emily P. Kleinbart
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Kelsey Martin
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Pietro M. Gentile
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Tae Won B. Kim
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
- Department of Orthopaedic Surgery, Cooper University Health Care, Camden, NJ, United States
| | - Sebastián L. Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, United States
- CORRESPONDENCE Sebastián L. Vega,
| |
Collapse
|
8
|
Wijekoon S, Sunaga T, Wang Y, Mwale C, Kim S, Okumura M. Pentosan polysulfate regulates hepcidin 1-facilitated formation and function of osteoclast derived from canine bone marrow. PLoS One 2022; 17:e0265596. [PMID: 35299233 PMCID: PMC8929557 DOI: 10.1371/journal.pone.0265596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/01/2022] [Indexed: 12/03/2022] Open
Abstract
Hepcidin which is the crucial regulator of iron homeostasis, produced in the liver in response to anemia, hypoxia, or inflammation. Recent studies have suggested that hepcidin and iron metabolism are involved in osteoporosis by inhibiting osteoblast function and promoting osteoclastogenesis. Pentosan polysulfate (PPS) is a heparin analogue and promising novel therapeutic for osteoarthritis (OA). This study was undertaken to determine whether PPS inhibits hepcidin-facilitated osteoclast (OC) differentiation and iron overload. Canine (n = 3) bone marrow mononuclear cells were differentiated to OC by macrophage colony-stimulating factor and receptor-activator of nuclear factor kappaB ligand with the treatment of hepcidin1 (200, 400, 800, 1200 nmol/L) and PPS (1, 5, 10, 20, 40 μg/mL). Differentiation and function of OC were accessed using tartrate-resistant acid phosphate staining and bone resorption assay while monitoring ferroportin1 (FPN1) and iron concentration by immunocytochemistry. Gene expression of OC for cathepsin K (CTK), matrix metallopeptidase-9, nuclear factor of activated-T-cells cytoplasmic 1 and FPN1 was examined. Hepcidin1 showed significant enhancement of OC number at 800 nmol/L (p<0.01). PPS impeded hepcidin-facilitated OC at 1, 5 and 10 μg/mL and reduction of resorption pits at 5 and 10 μg/mL (p< 0.01). All OC specific genes were downregulated with PPS, specifically in significant manner with CTK at higher concentrations. However, heparin induced FPN1 internalization and degradation was inhibited at higher concentrations of PPS while restoring iron-releasing capability of OC. We demonstrate for the first time that PPS is a novel-inhibitor of hepcidin-facilitated OC formation/function which might be beneficial for treatment of OA and osteoporosis.
Collapse
Affiliation(s)
- Suranji Wijekoon
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Sapporo, Hokkaido, Japan
| | - Takafumi Sunaga
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Sapporo, Hokkaido, Japan
| | - Yanlin Wang
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Sapporo, Hokkaido, Japan
| | - Carol Mwale
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Sapporo, Hokkaido, Japan
| | - Sangho Kim
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Sapporo, Hokkaido, Japan
| | - Masahiro Okumura
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
9
|
Abstract
Hypertrophic chondrocytes are the master regulators of endochondral ossification; however, their ultimate cell fates cells remain largely elusive due to their transient nature. Historically, hypertrophic chondrocytes have been considered as the terminal state of growth plate chondrocytes, which are destined to meet their inevitable demise at the primary spongiosa. Chondrocyte hypertrophy is accompanied by increased organelle synthesis and rapid intracellular water uptake, which serve as the major drivers of longitudinal bone growth. This process is delicately regulated by major signaling pathways and their target genes, including growth hormone (GH), insulin growth factor-1 (IGF-1), indian hedgehog (Ihh), parathyroid hormone-related protein (PTHrP), bone morphogenetic proteins (BMPs), sex determining region Y-box 9 (Sox9), runt-related transcription factors (Runx) and fibroblast growth factor receptors (FGFRs). Hypertrophic chondrocytes orchestrate endochondral ossification by regulating osteogenic-angiogenic and osteogenic-osteoclastic coupling through the production of vascular endothelial growth factor (VEGF), receptor activator of nuclear factor kappa-B ligand (RANKL) and matrix metallopeptidases-9/13 (MMP-9/13). Hypertrophic chondrocytes also indirectly regulate resorption of the cartilaginous extracellular matrix, by controlling formation of a special subtype of osteoclasts termed "chondroclasts". Notably, hypertrophic chondrocytes may possess innate potential for plasticity, reentering the cell cycle and differentiating into osteoblasts and other types of mesenchymal cells in the marrow space. We may be able to harness this unique plasticity for therapeutic purposes, for a variety of skeletal abnormalities and injuries. In this review, we discuss the morphological and molecular properties of hypertrophic chondrocytes, which carry out important functions during skeletal growth and regeneration.
Collapse
Affiliation(s)
- Shawn A Hallett
- University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Wanida Ono
- University of Michigan School of Dentistry, Ann Arbor, MI, USA
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| | - Noriaki Ono
- University of Michigan School of Dentistry, Ann Arbor, MI, USA
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA.
| |
Collapse
|
10
|
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
|
11
|
Bone targeting nanocarrier-assisted delivery of adenosine to combat osteoporotic bone loss. Biomaterials 2021; 273:120819. [PMID: 33892345 PMCID: PMC10108099 DOI: 10.1016/j.biomaterials.2021.120819] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 11/22/2022]
Abstract
Extracellular adenosine has been shown to play a key role in maintaining bone health and could potentially be used to treat bone loss. However, systemic administration of exogenous adenosine to treat bone disorders remains a challenge due to the ubiquitous presence of adenosine receptors in different organs and the short half-life of adenosine in circulation. Towards this, we have developed a bone-targeting nanocarrier and determined its potential for systemic administration of adenosine. The nanocarrier, synthesized via emulsion suspension photopolymerization, is comprised of hyaluronic acid (HA) copolymerized with phenylboronic acid (PBA), a moiety that can form reversible bonds with adenosine. The bone binding affinity of the nanocarrier was achieved by alendronate (Aln) conjugation. Nanocarriers functionalized with the alendronate (Aln-NC) showed a 45% higher accumulation in the mice vertebrae in vivo compared to those lacking alendronate molecules (NCs). Systemic administration of adenosine via bone-targeting nanocarriers (Aln-NC) attenuated bone loss in ovariectomized (OVX) mice. Furthermore, bone tissue of mice treated with adenosine-loaded Aln-NC displayed trabecular bone characteristics comparable to healthy controls as shown by microcomputed tomography, histochemical staining, bone labeling, and mechanical strength. Overall, our results demonstrate the use of a bone-targeting nanocarrier towards systemic administration of adenosine and its application in treating bone degenerative diseases such as osteoporosis.
Collapse
|
12
|
Hathaway-Schrader JD, Novince CM. Maintaining homeostatic control of periodontal bone tissue. Periodontol 2000 2021; 86:157-187. [PMID: 33690918 DOI: 10.1111/prd.12368] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alveolar bone is a unique osseous tissue due to the proximity of dental plaque biofilms. Periodontal health and homeostasis are mediated by a balanced host immune response to these polymicrobial biofilms. Dysbiotic shifts within dental plaque biofilms can drive a proinflammatory immune response state in the periodontal epithelial and gingival connective tissues, which leads to paracrine signaling to subjacent bone cells. Sustained chronic periodontal inflammation disrupts "coupled" osteoclast-osteoblast actions, which ultimately result in alveolar bone destruction. This chapter will provide an overview of alveolar bone physiology and will highlight why the oral microbiota is a critical regulator of alveolar bone remodeling. The ecology of dental plaque biofilms will be discussed in the context that periodontitis is a polymicrobial disruption of host homeostasis. The pathogenesis of periodontal bone loss will be explained from both a historical and current perspective, providing the opportunity to revisit the role of fibrosis in alveolar bone destruction. Periodontal immune cell interactions with bone cells will be reviewed based on our current understanding of osteoimmunological mechanisms influencing alveolar bone remodeling. Lastly, probiotic and prebiotic interventions in the oral microbiota will be evaluated as potential noninvasive therapies to support alveolar bone homeostasis and prevent periodontal bone loss.
Collapse
Affiliation(s)
- Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
13
|
Yu W, Zhong L, Yao L, Wei Y, Gui T, Li Z, Kim H, Holdreith N, Jiang X, Tong W, Dyment N, Liu XS, Yang S, Choi Y, Ahn J, Qin L. Bone marrow adipogenic lineage precursors promote osteoclastogenesis in bone remodeling and pathologic bone loss. J Clin Invest 2021; 131:140214. [PMID: 33206630 DOI: 10.1172/jci140214] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is maintained by coupled activities of bone-forming osteoblasts/osteocytes and bone-resorbing osteoclasts. Alterations in this relationship can lead to pathologic bone loss such as osteoporosis. It is well known that osteogenic cells support osteoclastogenesis via production of RANKL. Interestingly, our recently identified bone marrow mesenchymal cell population-marrow adipogenic lineage precursors (MALPs) that form a multidimensional cell network in bone-was computationally demonstrated to be the most interactive with monocyte-macrophage lineage cells through high and specific expression of several osteoclast regulatory factors, including RANKL. Using an adipocyte-specific Adipoq-Cre to label MALPs, we demonstrated that mice with RANKL deficiency in MALPs have a drastic increase in trabecular bone mass in long bones and vertebrae starting from 1 month of age, while their cortical bone appears normal. This phenotype was accompanied by diminished osteoclast number and attenuated bone formation at the trabecular bone surface. Reduced RANKL signaling in calvarial MALPs abolished osteolytic lesions after LPS injections. Furthermore, in ovariectomized mice, elevated bone resorption was partially attenuated by RANKL deficiency in MALPs. In summary, our studies identified MALPs as a critical player in controlling bone remodeling during normal bone metabolism and pathological bone loss in a RANKL-dependent fashion.
Collapse
Affiliation(s)
- Wei Yu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Gui
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Bone and Joint Surgery, Institute of Orthopedic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Ziqing Li
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hyunsoo Kim
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicholas Holdreith
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xi Jiang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wei Tong
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nathaniel Dyment
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - X Sherry Liu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Cawley KM, Bustamante-Gomez NC, Guha AG, MacLeod RS, Xiong J, Gubrij I, Liu Y, Mulkey R, Palmieri M, Thostenson JD, Goellner JJ, O'Brien CA. Local Production of Osteoprotegerin by Osteoblasts Suppresses Bone Resorption. Cell Rep 2020; 32:108052. [PMID: 32905775 PMCID: PMC7493998 DOI: 10.1016/j.celrep.2020.108052] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/15/2020] [Accepted: 07/29/2020] [Indexed: 11/21/2022] Open
Abstract
Osteoprotegerin (OPG) inhibits the ability of receptor activator of nuclear factor κB (NF-κB) ligand (RANKL) to stimulate the differentiation, activity, and survival of bone-resorbing osteoclasts. Genetic studies in mice show that osteocytes are an important source of RANKL, but the cellular sources of OPG are unclear. We use conditional deletion of Tnfrsf11b, which encodes OPG, from different cell populations to identify functionally relevant sources of OPG in mice. Deletion from B lymphocytes and osteocytes, two cell types commonly thought to supply OPG, has little or no impact on bone mass. By contrast, deletion of Tnfrsf11b from osteoblasts increases bone resorption and reduces bone mass to an extent similar to germline deletion, demonstrating that osteoblasts are an essential source of OPG. These results suggest that, in addition to producing new bone matrix, osteoblasts also play an active role in terminating the resorption phase of the bone remodeling cycle by suppressing RANKL activity.
Collapse
Affiliation(s)
- Keisha M Cawley
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy Cecile Bustamante-Gomez
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anveshi G Guha
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ryan S MacLeod
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jinhu Xiong
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Igor Gubrij
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yu Liu
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Robin Mulkey
- Division of Laboratory Animal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Michela Palmieri
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jeff D Thostenson
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Joseph J Goellner
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Charles A O'Brien
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.
| |
Collapse
|
15
|
Wang X, Yamauchi K, Mitsunaga T. A review on osteoclast diseases and osteoclastogenesis inhibitors recently developed from natural resources. Fitoterapia 2020; 142:104482. [PMID: 31954740 DOI: 10.1016/j.fitote.2020.104482] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/09/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022]
Abstract
Natural products have been investigated as potential candidates of novel therapeutics and play a crucial role in advanced medicinal drugs. Natural resources, including local medicinal plants (especially folk medicinal plants), animals, bacteria, and fungi have been used for more than a century, and are precious gifts from nature, providing potential medicines with high safety. Osteoclast-related diseases, such as osteoporosis, rheumatoid arthritis, Paget's disease, osteoclastoma, and periprosthetic osteolysis, are currently the most common reasons for bone inflammation, pain and fractures, resulting in low quality of life. However, the curative effects of current therapeutic drugs for these osteoclast-related diseases are limited, and long-term treatment is needed. Further, in severe cases, surgical treatments are necessary, which may cause unaffordable expenses and subsequent influences such as neuralgia, mental stress, and even development of cancer. Thus, safer inhibitors and potential drugs with enhanced curative effects and quick relief are needed to treat patients with osteoclast diseases. This review aims to introduce the main osteoclast-related diseases and some of the recently developed naturally sourced inhibitors against osteoclastogenesis, also it is desired to attract people's attention on using widely available natural resources for the evolution of new types of osteoclast inhibitors with minimal or no side-effects upon long-term treatments.
Collapse
Affiliation(s)
- Xiaoyu Wang
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | - Kosei Yamauchi
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | - Tohru Mitsunaga
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan.
| |
Collapse
|
16
|
Moore ER, Chen JC, Jacobs CR. Prx1-Expressing Progenitor Primary Cilia Mediate Bone Formation in response to Mechanical Loading in Mice. Stem Cells Int 2019; 2019:3094154. [PMID: 31814831 PMCID: PMC6877967 DOI: 10.1155/2019/3094154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/14/2019] [Accepted: 09/26/2019] [Indexed: 11/25/2022] Open
Abstract
Increases in mechanical loading can enhance the addition of new bone, altering geometry and density such that bones better withstand higher forces. Bone-forming osteoblasts have long been thought to originate from progenitors, but the exact source is yet to be identified. Previous studies indicate osteogenic precursors arise from Prx1-expressing progenitors during embryonic development and adult fracture repair. However, it is unknown whether this cell population is also a source for mechanically induced active osteoblasts. We first identified that Prx1 is expressed in skeletally mature mouse periosteum, a thin tissue covering the surface of the bone that is rich in osteoprogenitors. We then traced Prx1 progenitor lineage using a transgenic mouse model carrying both a Prx1-driven tamoxifen-inducible Cre and a ROSA-driven lacZ reporter gene. Cells that expressed Prx1 when compressive axial loading was applied were detected within the cortical bone days after stimulation, indicating osteocytes are of Prx1-expressing cell origin. In addition, we evaluated how these cells sense and respond to physical stimulation in vivo by disrupting their primary cilia, which are antenna-like sensory organelles known to enhance mechanical and chemical signaling kinetics. Although Prx1-driven primary cilium disruption did not affect osteoblast recruitment to the bone surface, the relative mineral apposition and bone formation rates were decreased by 53% and 34%, respectively. Thus, this cell population contributes to load-induced bone formation, and primary cilia are needed for a complete response. Interestingly, Prx1-expressing progenitors are easily extracted from periosteum and are perhaps an attractive alternative to marrow stem cells for bone tissue regeneration strategies.
Collapse
Affiliation(s)
- Emily R. Moore
- Department of Biomedical Engineering, Columbia University, 500 W 120th Street New York, NY 10027, USA
| | - Julia C. Chen
- Department of Biomedical Engineering, Columbia University, 500 W 120th Street New York, NY 10027, USA
| | - Christopher R. Jacobs
- Department of Biomedical Engineering, Columbia University, 500 W 120th Street New York, NY 10027, USA
| |
Collapse
|
17
|
Wang X, Batubara I, Yamauchi K, Mitsunaga T. Identification and structure-activity relationship (SAR) of chemical constituents from Daemonorops draco (Willd.) Blume and selected commercial flavonoids on anti-osteoclastogenesis activity. Fitoterapia 2019; 138:104280. [PMID: 31376421 DOI: 10.1016/j.fitote.2019.104280] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 01/28/2023]
Abstract
Osteoclastogenesis-related bone diseases including osteoporosis, rheumatoid arthritis, Paget's disease and periodontitis are worldwide occurred and cause severe health problems including bone fracture and bone cancer. However, A few studies have shown that Daemonorops draco (Willd.) Blume may decrease bone destruction and relieve bone cancer pain. In this research, we isolated and purified four known and two novel compounds from D. draco and investigated their anti-osteoclastogenesis activity using RAW264.7 cells. Among them, com.1 exhibited the most effective inhibitory activity on osteoclastogenesis with 78% inhibition at 10 μM and identified to be a novel natural flavan; and com.2 displayed a bit slighter inhibition (50% at 10 μM), indicating that the methylation of 7-hydroxyl group increased the anti-osteoclastogenesis activity. Moreover, nineteen commercial flavonoids were also performed in this study to investigate their inhibitory activity on osteoclastogenesis, and furtherly develop the SAR profile in flavonoid skeleton combined with the information of isolated compounds. Interestingly, the absence of substituents in B-ring and (3R)-hydroxyl group seems to play a crucial role in increasing anti-osteoclastogenesis activity.
Collapse
Affiliation(s)
- Xiaoyu Wang
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | - Irmanida Batubara
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Tropical Biopharmaca Research Center, Bogor Agriculture University, Bogor 16128, Indonesia
| | - Kosei Yamauchi
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | - Tohru Mitsunaga
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan.
| |
Collapse
|
18
|
A review of materials for managing bone loss in revision total knee arthroplasty. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109941. [PMID: 31500053 DOI: 10.1016/j.msec.2019.109941] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 06/14/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022]
Abstract
In 2014-2015, 61,421 total knee arthroplasties (TKAs) were performed in Canada; an increase of about 20% over 2000-2001. Revision total knee arthroplasties (rTKAs) accounted for 6.8% of TKAs performed between 2014 and 2015, and this is estimated to grow another 12% by 2025. rTKAs are typically more complicated than primary TKAs due to the significant loss of femoral and tibial bone stock. The escalating demand and limitations associated with total knee arthroplasty and their revision drives the development of novel treatments. A variety of materials have been utilized to facilitate regeneration of healthy bone around the site of a knee arthroplasty. The selection of these materials is based on the bone defect size and includes bone grafts, graft substitutes and cements. However, all these materials have certain disadvantages such as blood loss, disease transmission (bone grafts), inflammatory response, insufficient mechanical properties (bone graft substitutes) thermal necrosis and stress shielding (bone cement). Recently, the use of metal augments for large bone defects has attracted attention, however they can undergo fretting, corrosion, and stress shielding. All things considered, this review indicates the necessity of developing augments that have structural integrities and biodegradation rates similar to that of human bone. Therefore, the future of bone loss management may lie in fabricating novel bioactive glass augments as they can promote bone healing and implant stability and can degrade with time.
Collapse
|
19
|
Kim EJ, Kim HJ, Baik SW, Kim KH, Ryu SJ, Kim CH, Shin SW. Propofol promotes osteoclastic bone resorption by increasing DC-STAMP expression. J Dent Anesth Pain Med 2019; 18:349-359. [PMID: 30637345 PMCID: PMC6323039 DOI: 10.17245/jdapm.2018.18.6.349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 11/15/2022] Open
Abstract
Background Propofol is an intravenous anesthetic which has antioxidant effects due to its similarity in molecular structure to α-tocopherol. It has been reported that α-tocopherol increases osteoclast fusion and bone resorption. Here, we investigated the effects of propofol on signaling pathways of osteoclastogenic gene expression, as well as osteoclastogenesis and bone resorption using bone marrow-derived macrophages (BMMs). Methods BMMs were cultured with macrophage colony-stimulating factor (M-CSF) alone or M-CSF plus receptor activator of nuclear factor kappa B ligand (RANKL) in the presence of propofol (0-50 µM) for 4 days. Mature osteoclasts were stained for tartrate-resistant acid phosphatase (TRAP) and the numbers of TRAP-positive multinucleated osteoclasts were counted. To examine the resorption activities of osteoclasts, a bone resorption assay was performed. To identify the mechanism of action of propofol on the formation of multinucleated osteoclasts, we focused on dendritic cell-specific transmembrane protein (DC-STAMP), a protein essential for pre-osteoclastic cell fusion. Results Propofol increased the formation of TRAP-positive multinucleated osteoclasts. In addition, the bone resorption assay revealed that propofol increased the bone resorption area on dentin discs. The mRNA expression of DC-STAMP was upregulated most strongly in the presence of both RANKL and propofol. However, SB203580, a p38 inhibitor, significantly suppressed the propofol/RANKL-induced increase in mRNA expression of DC-STAMP. Conclusion We have demonstrated that propofol enhances osteoclast differentiation and maturation, and subsequently increases bone resorption. Additionally, we identified the regulatory pathway underlying osteoclast cell-cell fusion, which was enhanced by propofol through p38-mediated DC-STAMP expression.
Collapse
Affiliation(s)
- Eun-Jung Kim
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| | - Hyung Joon Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| | - Seong Wan Baik
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Kyung-Hoon Kim
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sie Jeong Ryu
- Department of Anesthesia and Pain Medicine, College of Medicine, Kosin University, Busan, Korea
| | - Cheul-Hong Kim
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| | - Sang-Wook Shin
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
20
|
Abstract
Bone is a crucial element of the skeletal-locomotor system, but also functions as an immunological organ that harbors hematopoietic stem cells (HSCs) and immune progenitor cells. Additionally, the skeletal and immune systems share a number of regulatory molecules, including cytokines and signaling molecules. Osteoimmunology was created as an interdisciplinary field to explore the shared molecules and interactions between the skeletal and immune systems. In particular, the importance of an inseparable link between the two systems has been highlighted by studies on the pathogenesis of rheumatoid arthritis (RA), in which pathogenic helper T cells induce the progressive destruction of multiple joints through aberrant expression of receptor activator of nuclear factor (NF)-κB ligand (RANKL). The conceptual bridge of osteoimmunology provides not only a novel framework for understanding these biological systems but also a molecular basis for the development of therapeutic approaches for diseases of bone and/or the immune system.
Collapse
Affiliation(s)
- Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
21
|
Ciliotherapy Treatments to Enhance Biochemically- and Biophysically-Induced Mesenchymal Stem Cell Osteogenesis: A Comparison Study. Cell Mol Bioeng 2018; 12:53-67. [PMID: 31719899 DOI: 10.1007/s12195-018-00561-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/07/2018] [Indexed: 01/12/2023] Open
Abstract
Introduction New approaches to treat osteoporosis have focused on promoting bone formation through the targeting of osteoblasts and their progenitors, mesenchymal stem cells (MSCs). The primary cilium is a singular cellular extension known to play an important role in biochemical and biophysical osteogenic induction of MSCs. Defects in ciliary structure have been associated with a plethora of diseases. Therefore targeting the cilium therapeutically (ciliotherapies) has emerged as a potential new treatment modality. Therefore, this study performed a comparison analysis on known ciliotherapies and their potential effects in mediating MSC osteogenic differentiation. Methods MSCs were treated with forskolin, lithium chloride (LiCl) or fenoldopam to investigate the effect on ciliogenesis and cilia-associated signalling. Moreover, both early and long term biochemical and biophysical (fluid shear) induced osteogenic differentiation was examined in terms of osteogenic gene expression and bone matrix deposition following each treatment. Results LiCl and fenoldopam were found to enhance MSC ciliogenesis to a similar degree. LiCl significantly altered hedgehog (HH) and Wnt signalling which was associated with inhibited osteogenic gene expression, while fenoldopam demonstrated enhanced early osteogenesis. Long term treatment with both ciliotherapies did not enhance osteogenesis, however LiCl had detrimental effects on cell viability. Intriguingly both ciliotherapies enhanced MSC mechanosensitivity as demonstrated by augmented osteogenic gene expression in response to fluid shear, which over longer durations resulted in enhanced matrix deposition per cell. Conclusions Therefore, ciliotherapies can be utilised to enhance MSC ciliogenesis resulting in enhanced mechanosensitivity, however, only fenoldopam is a viable ciliotherapeutic option to enhance MSC osteogenesis.
Collapse
|
22
|
Wijekoon HMS, Bwalya EC, Fang J, Kim S, Hosoya K, Okumura M. Inhibitory effects of sodium pentosan polysulfate on formation and function of osteoclasts derived from canine bone marrow. BMC Vet Res 2018; 14:152. [PMID: 29720166 PMCID: PMC5930774 DOI: 10.1186/s12917-018-1466-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 04/20/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Sodium pentosan polysulfate (NaPPS) was testified as a chondroprotective drug in with a detailed rationale of the disease-modifying activity. This study was undertaken to determine whether anti-osteoarthritis drug, NaPPS inhibited osteoclasts (OC) differentiation and function. Canine bone marrow mononuclear cells (n = 6) were differentiated to OC by maintaining with receptor activator of nuclear factor kappa B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) for up to 7 days with the treatment of NaPPS at concentration of 0, 0.2, 1 and 5 μg/mL. Differentiation and function of OC were accessed using tartrate-resistant acid phosphate (TRAP) staining and bone resorption assay, while monitoring actin ring formation. Invasion and colocalization patterns of fluorescence-labeled NaPPS with transcribed gene in OC were monitored. Gene expression of OC for cathepsin K (CTK), matrix metallopeptidase-9 (MMP-9), nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), c-Fos, activator protein-1(AP-1) and carbonic anhydrase II was examined using real-time PCR. RESULTS Significant inhibition of OC differentiation was evident at NaPPS concentration of 1 and 5 μg/mL (p < 0.05). In the presence of 0.2 to 5 μg/mL NaPPS, bone resorption was attenuated (p < 0.05), while 1 and 5 μg/mL NaPPS achieved significant reduction of actin ring formation. Intriguingly, fluorescence-labeled NaPPS invaded in to cytoplasm and nucleus while colocalizing with actively transcribed gene. Gene expression of CTK, MMP-9 and NFATc1 were significantly inhibited at 1 and 5 μg/mL (p < 0.05) of NaPPS whereas inhibition of c-Fos and AP-1 was identified only at concentration of 5 μg/mL (p < 0.05). CONCLUSIONS Taken together, all the results suggest that NaPPS is a novel inhibitor of RANKL and M-CSF-induced CTK, MMP-9, NFATc1, c-Fos, AP-1 upregulation, OC differentiation and bone resorption which might be a beneficial for treatment of inflammatory joint diseases and other bone diseases associated with excessive bone resorption.
Collapse
Affiliation(s)
- H. M. Suranji Wijekoon
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Surgery, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818 Japan
| | - Eugene C. Bwalya
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Surgery, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818 Japan
| | - Jing Fang
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Surgery, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818 Japan
| | - Sangho Kim
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Surgery, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818 Japan
| | - Kenji Hosoya
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Surgery, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818 Japan
| | - Masahiro Okumura
- Department of Veterinary Clinical Sciences, Laboratory of Veterinary Surgery, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818 Japan
| |
Collapse
|
23
|
|
24
|
Jeon HO, Choi IS, Yoon JY, Kim EJ, Yoon JU, Cho AR, Kim HJ, Kim CH. Effect of remifentanil on pre-osteoclast cell differentiation in vitro. J Dent Anesth Pain Med 2018; 18:9-17. [PMID: 29556554 PMCID: PMC5858013 DOI: 10.17245/jdapm.2018.18.1.9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 01/25/2018] [Accepted: 01/25/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The structure and function of bone tissue is maintained through a constant remodeling process, which is maintained by the balance between osteoblasts and osteoclasts. The failure of bone remodeling can lead to pathological conditions of bone structure and function. Remifentanil is currently used as a narcotic analgesic agent in general anesthesia and sedation. However, the effect of remifentanil on osteoclasts has not been studied. Therefore, we investigated the effect of remifentanil on pre-osteoclast (pre-OCs) differentiation and the mechanism of osteoclast differentiation in the absence of specific stimulus. METHODS Pre-OCs were obtained by culturing bone marrow-derived macrophages (BMMs) in osteoclastogenic medium for 2 days and then treated with various concentration of remifentanil. The mRNA expression of NFATc1 and c-fos was examined by using real-time PCR. We also examined the effect of remifentanil on the osteoclast-specific genes TRAP, cathepsin K, calcitonin receptor, and DC-STAMP. Finally, we examined the influence of remifentanil on the migration of pre-OCs by using the Boyden chamber assay. RESULTS Remifentanil increased pre-OC differentiation and osteoclast size, but did not affect the mRNA expression of NFATc1 and c-fos or significantly affect the expression of TRAP, cathepsin K, calcitonin receptor, and DC-STAMP. However, remifentanil increased the migration of pre-OCs. CONCLUSIONS This study suggested that remifentanil promotes the differentiation of pre-OCs and induces maturation, such as increasing osteoclast size. In addition, the increase in osteoclast size was mediated by the enhancement of pre-OC migration and cell fusion.
Collapse
Affiliation(s)
- Hyun-Ook Jeon
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| | - In-Seok Choi
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| | - Ji-Young Yoon
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| | - Eun-Jung Kim
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| | - Ji-Uk Yoon
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ah-Reum Cho
- Department of Anesthesia and Pain Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Hyung-Joon Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| | - Cheul-Hong Kim
- Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute, Yangsan, Republic of Korea
| |
Collapse
|
25
|
Lee DW, Kwon JY, Kim HK, Lee HJ, Kim ES, Kim HJ, Kim HJ, Lee HB. Propofol attenuates osteoclastogenesis by lowering RANKL/OPG ratio in mouse osteoblasts. Int J Med Sci 2018; 15:723-729. [PMID: 29910677 PMCID: PMC6001417 DOI: 10.7150/ijms.22713] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 04/12/2018] [Indexed: 11/29/2022] Open
Abstract
Bone remodeling plays an important role in the bone healing process; for example, following fracture. The relative ratio of the receptor activator of nuclear factor kappa B ligand (RANKL)/ osteoprotegerin (OPG) controls osteoclast differentiation, thereby playing a pivotal role in the regulation of bone remodeling. Propofol, a widely used anesthetic agent in orthopedic procedures, is considered to possess potential antioxidant properties owing to its structural similarity to α-tocopherol. Antioxidants are known to enhance bone healing. Accordingly, in the present study, we aimed to investigate osteoblastic differentiation and RANKL/OPG expression following propofol administration, in order to assess the potentially beneficial effects of this drug on the bone remodeling process, using calvarial primary osteoblasts from newborn mice. Calvarial pre-osteoblast cells were cultured in media containing clinically relevant concentrations of propofol, and cytotoxicity, effects on cell proliferation, osteogenic activity, and osteoclastogenesis were examined. The present findings indicated that propofol did not exert cytotoxic effects or alter cell proliferation in primary calvarial osteoblasts. Further, propofol did not affect osteoblast differentiation. The RANKL/OPG ratio was found to be decreased following propofol administration, and osteoclastogenesis was significantly reduced, indicating that propofol attenuated the osteoclastogenesis-supporting activity of osteoblasts. The results demonstrate that propofol, at clinically relevant concentrations, exerts beneficial effects on bone remodeling by attenuating osteoclastogenesis via suppression of the RANKL/OPG expression axis.
Collapse
Affiliation(s)
- Do-Won Lee
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jae-Young Kwon
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hae-Kyu Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hyeon-Jeong Lee
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Eun-Soo Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hyae-Jin Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hyung-Joon Kim
- Department of Oral Physiology, BK21 PLUS Project, and Institute of Translational Dental Sciences, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| | - Han-Bit Lee
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
26
|
Li R, Zhou Y, Cao Z, Liu L, Wang J, Chen Z, Xing W, Chen S, Bai J, Yuan W, Cheng T, Xu M, Yang FC, Zhao Z. TET2 Loss Dysregulates the Behavior of Bone Marrow Mesenchymal Stromal Cells and Accelerates Tet2 -/--Driven Myeloid Malignancy Progression. Stem Cell Reports 2017; 10:166-179. [PMID: 29290626 PMCID: PMC5768963 DOI: 10.1016/j.stemcr.2017.11.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 11/25/2017] [Accepted: 11/27/2017] [Indexed: 12/26/2022] Open
Abstract
TET2 is a methylcytosine dioxygenase that regulates cytosine hydroxymethylation. Although there are extensive data implicating a pivotal role of TET2 in hematopoietic stem/progenitor cells (HSPCs), the importance of TET2 in bone marrow mesenchymal stromal cells (BMSCs) remains unknown. In this study, we show that loss of TET2 in BMSCs increases cell proliferation and self-renewal and enhances osteoblast differentiation potential of BMSCs, which may in turn alter their behavior in supporting HSPC proliferation and differentiation. In addition, Tet2 loss alters BMSCs in promoting Tet2-deficiency-mediated myeloid malignancy progression. Tet2 loss in BMSCs also dysregulates hydroxylation of 5-methylcytosine (5mC) and the expression of genes that are key for BMSC proliferation and osteoblast differentiation, leading to alteration of biological characteristics in vivo. These results highlight the critical role of TET2 in the maintenance of BMSC functions and osteoblast differentiation and provide evidence that dysregulation of epigenetic modifiers in BMSCs contributes to the progression of myeloid malignancies.
Collapse
Affiliation(s)
- Rong Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Zeng Cao
- Department of Hematology and Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Lin Liu
- Department of Hematology and Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Jinhuan Wang
- Department of Oncology, The Second Affiliated Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zizhen Chen
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Wen Xing
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Shi Chen
- Sylvester Comprehensive Cancer Center, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jie Bai
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Mingjiang Xu
- Sylvester Comprehensive Cancer Center, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Feng-Chun Yang
- Sylvester Comprehensive Cancer Center, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Zhigang Zhao
- Department of Hematology and Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| |
Collapse
|
27
|
Le BQ, Nurcombe V, Cool SM, van Blitterswijk CA, de Boer J, LaPointe VLS. The Components of Bone and What They Can Teach Us about Regeneration. MATERIALS (BASEL, SWITZERLAND) 2017; 11:E14. [PMID: 29271933 PMCID: PMC5793512 DOI: 10.3390/ma11010014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/18/2022]
Abstract
The problem of bone regeneration has engaged both physicians and scientists since the beginning of medicine. Not only can bone heal itself following most injuries, but when it does, the regenerated tissue is often indistinguishable from healthy bone. Problems arise, however, when bone does not heal properly, or when new tissue is needed, such as when two vertebrae are required to fuse to stabilize adjacent spine segments. Despite centuries of research, such procedures still require improved therapeutic methods to be devised. Autologous bone harvesting and grafting is currently still the accepted benchmark, despite drawbacks for clinicians and patients that include limited amounts, donor site morbidity, and variable quality. The necessity for an alternative to this "gold standard" has given rise to a bone-graft and substitute industry, with its central conundrum: what is the best way to regenerate bone? In this review, we dissect bone anatomy to summarize our current understanding of its constituents. We then look at how various components have been employed to improve bone regeneration. Evolving strategies for bone regeneration are then considered.
Collapse
Affiliation(s)
- Bach Quang Le
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #6-06 Immunos, Singapore 138648, Singapore.
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #6-06 Immunos, Singapore 138648, Singapore.
| | - Simon McKenzie Cool
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #6-06 Immunos, Singapore 138648, Singapore.
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore 119288, Singapore.
| | - Clemens A van Blitterswijk
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Jan de Boer
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Vanessa Lydia Simone LaPointe
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
28
|
Wein MN. Parathyroid Hormone Signaling in Osteocytes. JBMR Plus 2017; 2:22-30. [PMID: 30283888 PMCID: PMC6124166 DOI: 10.1002/jbm4.10021] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 12/13/2022] Open
Abstract
Osteocytes are the most abundant cell type in bone and play a central role in orchestrating skeletal remodeling, in part by producing paracrine‐acting factors that in turn influence osteoblast and osteoclast activity. Recent evidence has indicated that osteocytes are crucial cellular targets of parathyroid hormone (PTH). Here, we will review the cellular and molecular mechanisms through which PTH influences osteocyte function. Two well‐studied PTH target genes in osteocytes are SOST and receptor activator of NF‐κB ligand (RANKL). The molecular mechanisms through which PTH regulates expression of these two crucial target genes will be discussed. Beyond SOST and RANKL, PTH/PTH‐related peptide (PTHrP) signaling in osteocytes may directly influence the way osteocytes remodel their perilacunar environment to influence bone homeostasis in a cell‐autonomous manner. Here, I will highlight novel, additional mechanisms used by PTH and PTHrP to modulate bone homeostasis through effects in osteocytes. © 2017 The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marc N Wein
- Endocrine Unit, Massachusetts General Hospital Harvard Medical School Boston MA USA
| |
Collapse
|
29
|
Xifra G, Moreno-Navarrete JM, Moreno M, Ricart W, Fernández-Real JM. Obesity status influences the relationship among serum osteocalcin, iron stores and insulin sensitivity. Clin Nutr 2017; 37:2091-2096. [PMID: 29050649 DOI: 10.1016/j.clnu.2017.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/15/2017] [Accepted: 10/02/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND & AIMS Increased iron stores significantly influence the clinical course of several chronic metabolic diseases. Recent studies have shown that iron overload decreases osteocalcin. We aimed to explore the relationship among osteocalcin, iron stores and insulin sensitivity. METHODS Extensive clinical and laboratory measurements, including serum ferritin, cross-linked C-telopeptide of type I collagen (CTX) and osteocalcin (OC) concentrations, were analyzed in 250 adult consecutive Caucasian men. Insulin sensitivity was evaluated through frequently sampled intravenous glucose tolerance tests with minimal model analysis. RESULTS Circulating serum ferritin were negatively associated with serum OC and CTX (p = 0.004 and p = 0.045 respectively). In all subjects as a whole, BMI and ferritin contributed to explain 5.2% of OC variance after controlling for age and smoking status. However, the association between OC and insulin sensitivity remained significant only in lean subjects (BMI < 25 kg/m2, r = 0.468; p = 0.006) whereas the link between serum ferritin concentration and OC and CTX were significant only in overweight/obese subjects (BMI ≥ 25 kg/m2, r = -0.229; p = 0.002 and r = -0.196; p = 0.008, respectively). CONCLUSIONS The association of circulating osteocalcin with parameters of insulin sensitivity and iron stores were dependent on obesity status. Increased iron stores could contribute to the detrimental metabolic effects of overweight and obesity on bone.
Collapse
Affiliation(s)
- Gemma Xifra
- Department of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IdIBGi), CIBER de la Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IdIBGi), CIBER de la Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - María Moreno
- Department of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IdIBGi), CIBER de la Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - Wifredo Ricart
- Department of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IdIBGi), CIBER de la Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition (UDEN), Institut d'Investigació Biomédica de Girona (IdIBGi), CIBER de la Fisiopatología de la Obesidad y la Nutrición (CIBERobn, CB06/03/0010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain.
| |
Collapse
|
30
|
Okamoto K, Nakashima T, Shinohara M, Negishi-Koga T, Komatsu N, Terashima A, Sawa S, Nitta T, Takayanagi H. Osteoimmunology: The Conceptual Framework Unifying the Immune and Skeletal Systems. Physiol Rev 2017; 97:1295-1349. [DOI: 10.1152/physrev.00036.2016] [Citation(s) in RCA: 369] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.
Collapse
Affiliation(s)
- Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Tomoki Nakashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masahiro Shinohara
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takako Negishi-Koga
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Noriko Komatsu
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Asuka Terashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Shinichiro Sawa
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takeshi Nitta
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
31
|
Zhu YL, Wang S, Ding DG, Xu L, Zhu HT. miR‑217 inhibits osteogenic differentiation of rat bone marrow‑derived mesenchymal stem cells by binding to Runx2. Mol Med Rep 2017; 15:3271-3277. [PMID: 28339007 DOI: 10.3892/mmr.2017.6349] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/03/2017] [Indexed: 11/06/2022] Open
Abstract
The elucidation of the underlying molecular mechanisms regulating the osteogenic differentiation of bone marrow‑derived mesenchymal stem cells (BMSCs) is of great importance in improving the treatment of bone‑associated diseases. MicroRNAs (miRNAs) have been proven to regulate the osteogenic differentiation of BMSCs. The present study investigated the role of miR‑217 in the osteogenic differentiation of rat BMSCs. It was observed that miR‑217 expression levels were downregulated during the process of osteogenic differentiation. Subsequently, a dual‑luciferase reporter gene assay demonstrated that miR‑217 targets a putative binding site in the 3'‑untranslated region of the runt related transcription factor 2 (Runx2) gene, which is a key transcription factor for osteogenesis. It was then demonstrated that overexpression of miR‑217 attenuated the osteogenesis of BMSCs and downregulated the expression of Runx2, whereas inhibition of miR‑217 promoted osteoblastic differentiation and upregulated Runx2 expression. Furthermore, the extracellular signal‑regulated kinase (ERK) and p38 mitogen‑activated protein kinase (p38 MAPK) signaling pathways were investigated during osteogenic induction, and the data indicated that miR‑217 may exert a negative effect on the osteogenic differentiation of BMSCs through alteration of ERK and p38 MAPK phosphorylation. The present study therefore concluded that miR‑217 functions as a negative regulator of BMSC osteogenic differentiation via the inhibition of Runx2 expression, and the underlying molecular mechanisms may partially be attributed to mediation by the ERK and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Yu-Long Zhu
- Department of Orthopedics, Sheyang County People's Hospital, Yancheng, Jiangsu 224300, P.R. China
| | - Shui Wang
- Department of Orthopedics, Sheyang County People's Hospital, Yancheng, Jiangsu 224300, P.R. China
| | - De-Gang Ding
- Department of Orthopedics, Sheyang County People's Hospital, Yancheng, Jiangsu 224300, P.R. China
| | - Liang Xu
- Department of Orthopedics, Sheyang County People's Hospital, Yancheng, Jiangsu 224300, P.R. China
| | - Hai-Tao Zhu
- Department of Orthopedics, Sheyang County People's Hospital, Yancheng, Jiangsu 224300, P.R. China
| |
Collapse
|
32
|
Abstract
Osteoporosis is a condition causing significant morbidity and mortality in the elderly population worldwide. Age-related testosterone deficiency is the most important factor of bone loss in elderly men. Androgen can influence bone health by binding to androgen receptors directly or to estrogen receptors (ERs) indirectly via aromatization to estrogen. This review summarized the direct and indirect effects of androgens on bone derived from in vitro, in vivo, and human studies. Cellular studies showed that androgen stimulated the proliferation of preosteoblasts and differentiation of osteoblasts. The converted estrogen suppressed osteoclast formation and resorption activity by blocking the receptor activator of nuclear factor k-B ligand pathway. In animal studies, activation of androgen and ERα, but not ERβ, was shown to be important in acquisition and maintenance of bone mass. Human epidemiological studies demonstrated a significant relationship between estrogen and testosterone in bone mineral density and fracture risk, but the relative significance between the two remained debatable. Human experimental studies showed that estrogen was needed in suppressing bone resorption, but both androgen and estrogen were indispensable for bone formation. As a conclusion, maintaining optimal level of androgen is essential in preventing osteoporosis and its complications in elderly men.
Collapse
Affiliation(s)
- Nur-Vaizura Mohamad
- Department of Pharmacology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lampur, Malaysia
| | - Ima-Nirwana Soelaiman
- Department of Pharmacology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lampur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lampur, Malaysia
| |
Collapse
|
33
|
Zimmer A, Otte DM, Bilkei-Gorzo A, Armin SM, Bab I. Behavioural changes induced by a conditional disruption of bone formation. J Basic Clin Physiol Pharmacol 2016; 27:203-7. [PMID: 26913456 DOI: 10.1515/jbcpp-2015-0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 01/02/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND It has been shown that the brain regulates bone remodelling through sympathetic and parasympathetic nerve fibres. However, it is unclear if signals from the skeleton also influence brain functions and animal behaviours. METHODS Bone formation was conditionally disrupted by daily injections of aciclovir (10 mg/kg) to transgenic mice expressing a herpes-simplex-virus thymidine kinase under the control of the osteoblast-specific promoter of the Bglap gene. Behavioural studies were conducted after 10 weeks of treatment. RESULTS Transgenic mice receiving aciclovir injections showed a reduced number of osteoblasts with a concomitantly reduced trabecular bone volume density, when compared to wild-type controls that were treated identically. The general health of the animals was not severely affected, as indicated by a similar increase in body weight, similar activity profiles and similar social behaviours. However, transgenic mice showed significantly increased despair behaviour and increased adrenal gland weights. CONCLUSIONS Specific animal behaviours can be modulated by a selective disruption of bone formation. The increased despair behaviour observed in transgenic animals indicates that these animals may be more prone to depression-related phenotypes. These findings are important in the context of the well-established clinical association between depression and reduced bone mass.
Collapse
|
34
|
Preclinical animal models of multiple myeloma. BONEKEY REPORTS 2016; 5:772. [PMID: 26909147 DOI: 10.1038/bonekey.2015.142] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 11/30/2015] [Indexed: 01/19/2023]
Abstract
Multiple myeloma is an incurable plasma-cell malignancy characterized by osteolytic bone disease and immunosuppression. Murine models of multiple myeloma and myeloma bone disease are critical tools for an improved understanding of the pathogenesis of the disease and the development of novel therapeutic strategies. This review will cover commonly used immunocompetent and xenograft models of myeloma, describing the advantages and disadvantages of each model system. In addition, this review provides detailed protocols for establishing systemic and local models of myeloma using both murine and human myeloma cell lines.
Collapse
|
35
|
The Modulatory Effects of Mesenchymal Stem Cells on Osteoclastogenesis. Stem Cells Int 2015; 2016:1908365. [PMID: 26823668 PMCID: PMC4707367 DOI: 10.1155/2016/1908365] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/21/2015] [Indexed: 12/14/2022] Open
Abstract
The effect of mesenchymal stem cells (MSCs) on bone formation has been extensively demonstrated through several in vitro and in vivo studies. However, few studies addressed the effect of MSCs on osteoclastogenesis and bone resorption. Under physiological conditions, MSCs support osteoclastogenesis through producing the main osteoclastogenic cytokines, RANKL and M-CSF. However, during inflammation, MSCs suppress osteoclast formation and activity, partly via secretion of the key anti-osteoclastogenic factor, osteoprotegerin (OPG). In vitro, co-culture of MSCs with osteoclasts in the presence of high concentrations of osteoclast-inducing factors might reflect the in vivo inflammatory pathology and prompt MSCs to exert an osteoclastogenic suppressive effect. MSCs thus seem to have a dual effect, by stimulating or inhibiting osteoclastogenesis, depending on the inflammatory milieu. This effect of MSCs on osteoclast formation seems to mirror the effect of MSCs on other immune cells, and may be exploited for the therapeutic potential of MSCs in bone loss associated inflammatory diseases.
Collapse
|
36
|
Hamed SA. Markers of bone turnover in patients with epilepsy and their relationship to management of bone diseases induced by antiepileptic drugs. Expert Rev Clin Pharmacol 2015; 9:267-286. [PMID: 26589104 DOI: 10.1586/17512433.2016.1123617] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Data from cross-sectional and prospective studies revealed that patients with epilepsy and on long-term treatment with antiepileptic drugs (AEDs) are at increased risk for metabolic bone diseases. Bone diseases were reported in about 50% of patients on AEDs. Low bone mineral density, osteopenia/osteoporosis, osteomalacia, rickets, altered concentration of bone turnover markers and fractures were reported with phenobarbital, phenytoin, carbamazepine, valproate, oxcarbazepine and lamotrigine. The mechanisms for AEDs-induced bone diseases are heterogeneous and include hypovitaminosis D, hypocalcemia and direct acceleration of bone loss and/or reduction of bone formation. This article reviews the evidence, predictors and mechanisms of AEDs-induced bone abnormalities and its clinical implications. For patients on AEDs, regular monitoring of bone health is recommended. Prophylactic administration of calcium and vitamin D is recommended for all patients. Treatment doses of calcium and vitamin D and even anti-resorptive drug therapy are reserved for patients at high risk of pathological fracture.
Collapse
Affiliation(s)
- Sherifa A Hamed
- a Department of Neurology and Psychiatry , Assiut University Hospital , Assiut , Egypt
| |
Collapse
|
37
|
Hamed SA. Markers of bone turnover in patients with epilepsy and their relationship to management of bone diseases induced by antiepileptic drugs. Expert Rev Clin Pharmacol 2015; 9:267-286. [PMID: 26589104 DOI: org/10.1586/17512433.2016.1123617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Data from cross-sectional and prospective studies revealed that patients with epilepsy and on long-term treatment with antiepileptic drugs (AEDs) are at increased risk for metabolic bone diseases. Bone diseases were reported in about 50% of patients on AEDs. Low bone mineral density, osteopenia/osteoporosis, osteomalacia, rickets, altered concentration of bone turnover markers and fractures were reported with phenobarbital, phenytoin, carbamazepine, valproate, oxcarbazepine and lamotrigine. The mechanisms for AEDs-induced bone diseases are heterogeneous and include hypovitaminosis D, hypocalcemia and direct acceleration of bone loss and/or reduction of bone formation. This article reviews the evidence, predictors and mechanisms of AEDs-induced bone abnormalities and its clinical implications. For patients on AEDs, regular monitoring of bone health is recommended. Prophylactic administration of calcium and vitamin D is recommended for all patients. Treatment doses of calcium and vitamin D and even anti-resorptive drug therapy are reserved for patients at high risk of pathological fracture.
Collapse
Affiliation(s)
- Sherifa A Hamed
- a Department of Neurology and Psychiatry , Assiut University Hospital , Assiut , Egypt
| |
Collapse
|
38
|
Chen JC, Hoey DA, Chua M, Bellon R, Jacobs CR. Mechanical signals promote osteogenic fate through a primary cilia-mediated mechanism. FASEB J 2015; 30:1504-11. [PMID: 26675708 DOI: 10.1096/fj.15-276402] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/08/2015] [Indexed: 01/21/2023]
Abstract
It has long been suspected, but never directly shown, that bone formed to accommodate an increase in mechanical loading is related to the creation of osteoblasts from skeletal stem cells. Indeed, biophysical stimuli potently regulate osteogenic lineage commitmentin vitro In this study, we transplanted bone marrow cells expressing green fluorescent protein, to enable lineage tracing, and subjected mice to a biophysical stimulus, to elicit a bone-forming response. We detected cells derived from transplanted progenitors embedded within the bone matrix near active bone-forming surfaces in response to loading, demonstrating for the first time, that mechanical signals enhance the homing and attachment of bone marrow cells to bone surfaces and the commitment to an osteogenic lineage of these cellsin vivo Furthermore, we used an inducible Cre/Lox recombination system to delete kinesin family member 3A (Kif3a), a gene that is essential for primary cilia formation, at will in transplanted cells and their progeny, regardless of which tissue may have incorporated them. Disruption of the mechanosensing organelle, the primary cilium in a progenitor population, significantly decreased the amount of bone formed in response to mechanical stimulation. The collective results of our study directly demonstrate that, in a novel experimental stem cell mechanobiology model, mechanical signals enhance osteogenic lineage commitmentin vivoand that the primary cilium contributes to this process.-Chen, J. C., Hoey, D. A., Chua, M., Bellon, R., Jacobs, C. R. Mechanical signals promote osteogenic fate through a primary cilia-mediated mechanism.
Collapse
Affiliation(s)
- Julia C Chen
- *Department of Biomedical Engineering and Department of Chemical Engineering, Columbia University, New York, New York, USA; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, and Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Aeronautical, and Biomedical Engineering, Centre for Applied Biomedical Engineering Research, Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; and Department of Biotechnology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David A Hoey
- *Department of Biomedical Engineering and Department of Chemical Engineering, Columbia University, New York, New York, USA; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, and Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Aeronautical, and Biomedical Engineering, Centre for Applied Biomedical Engineering Research, Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; and Department of Biotechnology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mardonn Chua
- *Department of Biomedical Engineering and Department of Chemical Engineering, Columbia University, New York, New York, USA; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, and Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Aeronautical, and Biomedical Engineering, Centre for Applied Biomedical Engineering Research, Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; and Department of Biotechnology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raymond Bellon
- *Department of Biomedical Engineering and Department of Chemical Engineering, Columbia University, New York, New York, USA; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, and Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Aeronautical, and Biomedical Engineering, Centre for Applied Biomedical Engineering Research, Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; and Department of Biotechnology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher R Jacobs
- *Department of Biomedical Engineering and Department of Chemical Engineering, Columbia University, New York, New York, USA; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, and Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Aeronautical, and Biomedical Engineering, Centre for Applied Biomedical Engineering Research, Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; and Department of Biotechnology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
39
|
Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD. The effects of tumour necrosis factor-α on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes. J Periodontal Res 2015; 51:549-66. [DOI: 10.1111/jre.12339] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Affiliation(s)
- K. Algate
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - D. R. Haynes
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - P. M. Bartold
- School of Dentistry; University of Adelaide; Adelaide SA Australia
| | - T. N. Crotti
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - M. D. Cantley
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
- Myeloma Research Laboratory; University of Adelaide; Adelaide SA Australia
| |
Collapse
|
40
|
Goldring SR. The osteocyte: key player in regulating bone turnover. RMD Open 2015; 1:e000049. [PMID: 26557372 PMCID: PMC4632148 DOI: 10.1136/rmdopen-2015-000049] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 02/20/2015] [Accepted: 02/25/2015] [Indexed: 12/31/2022] Open
Abstract
Osteocytes are the most abundant cell type in bone and are distributed throughout the mineralised bone matrix forming an interconnected network that ideally positions them to sense and to respond to local biomechanical and systemic stimuli to regulate bone remodelling and adaptation. The adaptive process is dependent on the coordinated activity of osteoclasts and osteoblasts that form a so called bone multicellular unit that remodels cortical and trabecular bone through a process of osteoclast-mediated bone resorption, followed by a phase of bone formation mediated by osteoblasts. Osteocytes mediate their effects on bone remodelling via both cell–cell interactions with osteoclasts and osteoblasts, but also via signaling through the release of soluble mediators. The remodelling process provides a mechanism for adapting the skeleton to local biomechanical factors and systemic hormonal influences and for replacing bone that has undergone damage from repetitive mechanical loading.
Collapse
Affiliation(s)
- Steven R Goldring
- Research Division , Hospital for Special Surgery, Weill Cornell Medical College , New York, New York , USA
| |
Collapse
|
41
|
Abstract
After it was proposed that the osteoblast lineage controlled the formation of osteoclasts, cell culture methods were developed that established this to be the case. Evidence was obtained that cytokines and hormones that promote osteoclast formation act first on osteoblast lineage cells to promote the production of a membrane-bound regulator of osteoclastogenesis. This proved to be receptor activator of NF-kB ligand (RANKL) a member of the tumor necrosis factor ligand family that acts upon its receptor RANK in the hematopoietic lineage, with interaction restricted by a decoy soluble receptor osteoprotegerin (OPG), also a product of the osteoblast lineage. The physiological roles of these factors were established through genetic and pharmacological studies, have led to a new physiology of bone, with complete revision of older ideas over the last 15 years, ultimately leading to the development of new pharmaceutical agents for bone disease.
Collapse
Affiliation(s)
- T John Martin
- Department of Medicine at St. Vincent's Hospital, St. Vincent's Institute of Medical Research and The University of Melbourne, 9 Princes St, Fitzroy, VIC, 3065, Australia,
| | | |
Collapse
|
42
|
Ha J, Hwang JH, Kwon SG, Park DH, Kim TW, Kang DG, Kang KH, Kim IS, Kim CW. MSK1 regulates RANKL-induced NFATc1 expression through CREB and c-Fos. J Biomed Res 2015. [DOI: 10.12729/jbr.2015.16.2.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
43
|
Omori A, Yoshimura Y, Deyama Y, Suzuki K. Rosmarinic acid and arbutin suppress osteoclast differentiation by inhibiting superoxide and NFATc1 downregulation in RAW 264.7 cells. Biomed Rep 2015; 3:483-490. [PMID: 26171153 DOI: 10.3892/br.2015.452] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/23/2015] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the effect of the natural polyphenols, rosmarinic acid and arbutin, on osteoclast differentiation in RAW 264.7 cells. Rosmarinic acid and arbutin suppressed osteoclast differentiation and had no cytotoxic effect on osteoclast precursor cells. Rosmarinic acid and arbutin inhibited superoxide production in a dose-dependent manner. mRNA expression of the master regulator of osteoclastogenesis, nuclear factor of activated T cells cytoplasmic 1 (NFATc1) and the osteoclast marker genes, matrix metalloproteinase-9, tartrate-resistant acid phosphatase and cathepsin-K, decreased following treatments with rosmarinic acid and arbutin. Furthermore, resorption activity decreased with the number of osteoclasts. These results suggest that rosmarinic acid and arbutin may be useful for the prevention and treatment of bone diseases, such as osteoporosis, through mechanisms involving inhibition of superoxide and downregulation of NFATc1.
Collapse
Affiliation(s)
- Akina Omori
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Yoshitaka Yoshimura
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Yoshiaki Deyama
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Kuniaki Suzuki
- Department of Molecular Cell Pharmacology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| |
Collapse
|
44
|
Bauer O, Sharir A, Kimura A, Hantisteanu S, Takeda S, Groner Y. Loss of osteoblast Runx3 produces severe congenital osteopenia. Mol Cell Biol 2015; 35:1097-109. [PMID: 25605327 PMCID: PMC4355527 DOI: 10.1128/mcb.01106-14] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/07/2014] [Accepted: 12/30/2014] [Indexed: 11/20/2022] Open
Abstract
Congenital osteopenia is a bone demineralization condition that is associated with elevated fracture risk in human infants. Here we show that Runx3, like Runx2, is expressed in precommitted embryonic osteoblasts and that Runx3-deficient mice develop severe congenital osteopenia. Runx3-deficient osteoblast-specific (Runx3(fl/fl)/Col1α1-cre), but not chondrocyte-specific (Runx3(fl/fl)/Col1α2-cre), mice are osteopenic. This demonstrates that an osteoblastic cell-autonomous function of Runx3 is required for proper osteogenesis. Bone histomorphometry revealed that decreased osteoblast numbers and reduced mineral deposition capacity in Runx3-deficient mice cause this bone formation deficiency. Neonatal bone and cultured primary osteoblast analyses revealed a Runx3-deficiency-associated decrease in the number of active osteoblasts resulting from diminished proliferation and not from enhanced osteoblast apoptosis. These findings are supported by Runx3-null culture transcriptome analyses showing significant decreases in the levels of osteoblastic markers and increases in the levels of Notch signaling components. Thus, while Runx2 is mandatory for the osteoblastic lineage commitment, Runx3 is nonredundantly required for the proliferation of these precommitted cells, to generate adequate numbers of active osteoblasts. Human RUNX3 resides on chromosome 1p36, a region that is associated with osteoporosis. Therefore, RUNX3 might also be involved in human bone mineralization.
Collapse
Affiliation(s)
- Omri Bauer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Amnon Sharir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ayako Kimura
- Department of Orthopedics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shay Hantisteanu
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shu Takeda
- Department of Orthopedics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoram Groner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
45
|
Granito RN, Bouleftour W, Sabido O, Lescale C, Thomas M, Aubin JE, Goodhardt M, Vico L, Malaval L. Absence of Bone Sialoprotein (BSP) Alters Profoundly Hematopoiesis and Upregulates Osteopontin. J Cell Physiol 2015; 230:1342-51. [DOI: 10.1002/jcp.24877] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/05/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Renata neves Granito
- INSERM U1059/LBTO; Université Jean Monnet-Université de Lyon; Saint-Étienne France
| | - Wafa Bouleftour
- INSERM U1059/LBTO; Université Jean Monnet-Université de Lyon; Saint-Étienne France
| | - Odile Sabido
- Centre Commun de Cytométrie en Flux; Faculté de Médecine; Université Jean Monnet - Université de Lyon; Saint-Étienne France
| | - Chloé Lescale
- INSERM UMRS940, Institut Universitaire d'Hématologie; Université Paris 7 Denis Diderot; Paris France
| | - Mireille Thomas
- INSERM U1059/LBTO; Université Jean Monnet-Université de Lyon; Saint-Étienne France
| | - Jane E. Aubin
- Department of Molecular Genetics; University of Toronto; Toronto Ontario Canada
| | - Michèle Goodhardt
- INSERM UMRS940, Institut Universitaire d'Hématologie; Université Paris 7 Denis Diderot; Paris France
| | - Laurence Vico
- INSERM U1059/LBTO; Université Jean Monnet-Université de Lyon; Saint-Étienne France
| | | |
Collapse
|
46
|
Mechanically stimulated bone cells secrete paracrine factors that regulate osteoprogenitor recruitment, proliferation, and differentiation. Biochem Biophys Res Commun 2015; 459:118-23. [PMID: 25721667 DOI: 10.1016/j.bbrc.2015.02.080] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/13/2015] [Indexed: 01/20/2023]
Abstract
Bone formation requires the recruitment, proliferation and osteogenic differentiation of mesenchymal progenitors. A potent stimulus driving this process is mechanical loading, yet the signalling mechanisms underpinning this are incompletely understood. The objective of this study was to investigate the role of the mechanically-stimulated osteocyte and osteoblast secretome in coordinating progenitor contributions to bone formation. Initially osteocytes (MLO-Y4) and osteoblasts (MC3T3) were mechanically stimulated for 24 hrs and secreted factors within the conditioned media were collected and used to evaluate mesenchymal stem cell (MSC) and osteoblast recruitment, proliferation and osteogenesis. Paracrine factors secreted by mechanically stimulated osteocytes significantly enhanced MSC migration, proliferation and osteogenesis and furthermore significantly increased osteoblast migration and proliferation when compared to factors secreted by statically cultured osteocytes. Secondly, paracrine factors secreted by mechanically stimulated osteoblasts significantly enhanced MSC migration but surprisingly, in contrast to the osteocyte secretome, inhibited MSC proliferation when compared to factors secreted by statically cultured osteoblasts. A similar trend was observed in osteoblasts. This study provides new information on mechanically driven signalling mechanisms in bone and highlights a contrasting secretome between cells at different stages in the bone lineage, furthering our understanding of loading-induced bone formation and indirect biophysical regulation of osteoprogenitors.
Collapse
|
47
|
Sapir-Koren R, Livshits G. Osteocyte control of bone remodeling: is sclerostin a key molecular coordinator of the balanced bone resorption-formation cycles? Osteoporos Int 2014; 25:2685-700. [PMID: 25030653 DOI: 10.1007/s00198-014-2808-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 07/02/2014] [Indexed: 12/22/2022]
Abstract
Osteocytes, entrapped within a newly mineralized bone matrix, possess a unique cellular identity due to a specialized morphology and a molecular signature. These features endow them to serve as a bone response mechanism for mechanical stress in their microenvironment. Sclerostin, a primarily osteocyte product, is widely considered as a mechanotranduction key molecule whose expression is suppressed by mechanical loading, or it is induced by unloading. This review presents a model suggesting that sclerostin is major mediator for integrating mechanical, local, and hormonal signals, sensed by the osteocytes, in controlling the remodeling apparatus. This central role is achieved through interplay between two opposing mechanisms: (1) unloading-induced high sclerostin levels, which antagonize Wnt-canonical-β-catenin signaling in osteocytes and osteoblasts, permitting simultaneously Wnt-noncanonical and/or other pathways in osteocytes and osteoclasts, directed at bone resorption; (2) mechanical loading results in low sclerostin levels, activation of Wnt-canonical signaling, and bone formation. Therefore, adaptive bone remodeling occurring at a distinct bone compartment is orchestrated by altered sclerostin levels, which regulate the expression of the other osteocyte-specific proteins, such as RANKL, OPG, and proteins encoded by "mineralization-related genes" (DMP1, PHEX, and probably FGF23). For example, under specific terms, sclerostin regulates differential RANKL and OPG production, and creates a dynamic RANKL/OPG ratio, leading either to bone formation or resorption. It also controls the expression of PHEX, DMP1, and most likely FGF23, leading to either bone matrix mineralization or its inhibition. Such opposing up- or down-regulation of remodeling phases allows osteocytes to function as an "external unit", ensuring transition from bone resorption to bone formation.Mini Abstract: The osteocyte network plays a central role in directing bone response either to mechanical loading, or to unloading, leading correspondingly to bone formation or resorption. This review shows a key role of the osteocyte-produced sclerostin as a major mediator of the molecular mechanisms involved in the process of adaptive bone remodeling.
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
|
48
|
Sims NA, Vrahnas C. Regulation of cortical and trabecular bone mass by communication between osteoblasts, osteocytes and osteoclasts. Arch Biochem Biophys 2014; 561:22-8. [DOI: 10.1016/j.abb.2014.05.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/15/2014] [Accepted: 05/18/2014] [Indexed: 12/11/2022]
|
49
|
Ng AH, Willett TL, Alman BA, Grynpas MD. Development, validation and characterization of a novel mouse model of Adynamic Bone Disease (ABD). Bone 2014; 68:57-66. [PMID: 25111968 DOI: 10.1016/j.bone.2014.07.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 11/29/2022]
Abstract
The etiology of Adynamic Bone Disease (ABD) is poorly understood but the hallmark of ABD is a lack of bone turnover. ABD occurs in renal osteodystrophy (ROD) and is suspected to occur in elderly patients on long-term anti-resorptive therapy. A major clinical concern of ABD is diminished bone quality and an increased fracture risk. To our knowledge, experimental animal models for ABD other than ROD-ABD have not been developed or studied. The objectives of this study were to develop a mouse model of ABD without the complications of renal ablation, and to characterize changes in bone quality in ABD relative to controls. To re-create the adynamic bone condition, 4-month old female Col2.3Δtk mice were treated with ganciclovir to specifically ablate osteoblasts, and pamidronate was used to inhibit osteoclastic resorption. Four groups of animals were used to characterize bone quality in ABD: Normal bone controls, No Formation controls, No Resorption controls, and an Adynamic group. After a 6-week treatment period, the animals were sacrificed and the bones were harvested for analyses. Bone quality assessments were conducted using established techniques including bone histology, quantitative backscattered electron imaging (qBEI), dual energy X-ray absorptiometry (DXA), microcomputed tomography (microCT), and biomechanical testing. Histomorphometry confirmed osteoblast-related hallmarks of ABD in our mouse model. Bone formation was near complete suppression in the No Formation and Adynamic specimens. Inhibition of bone resorption in the Adynamic group was confirmed by tartrate-resistant acid phosphatase (TRAP) stain. Normal bone mineral density and architecture were maintained in the Adynamic group, whereas the No Formation group showed a reduction in bone mineral content and trabecular thickness relative to the Adynamic group. As expected, the No Formation group had a more hypomineralized profile and the Adynamic group had a higher mean mineralization profile that is similar to suppressed bone turnover in human. This data confirms successful replication of the adynamic bone condition in a mouse without the complication of renal ablation. Our approach is the first model of ABD that uses pharmacological manipulation in a transgenic mouse to mimic the bone cellular dynamics observed in the human ABD condition. We plan to use our mouse model to investigate the adynamic bone condition in aging and to study changes to bone quality and fracture risk as a consequence of over-suppressed bone turnover.
Collapse
Affiliation(s)
- Adeline H Ng
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Thomas L Willett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin A Alman
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Marc D Grynpas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
50
|
Abou-Khalil R, Colnot C. Cellular and molecular bases of skeletal regeneration: what can we learn from genetic mouse models? Bone 2014; 64:211-21. [PMID: 24709685 DOI: 10.1016/j.bone.2014.03.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 03/19/2014] [Accepted: 03/26/2014] [Indexed: 10/25/2022]
Abstract
Although bone repairs through a very efficient regenerative process in 90% of the patients, many factors can cause delayed or impaired healing. To date, there are no reliable biological parameters to predict or diagnose bone repair defects. Orthopedic surgeons mostly base their diagnoses on radiographic analyses. With the recent progress in our understanding of the bone repair process, new methods may be envisioned. Animal models have allowed us to define the key steps of bone regeneration and the biological and mechanical factors that may influence bone healing in positive or negative ways. Most importantly, small animal models such as mice have provided powerful tools to apprehend the genetic bases of normal and impaired bone healing. The current review presents a state of the art of the genetically modified mouse models that have advanced our understanding of the cellular and molecular components of bone regeneration and repair. The review illustrates the use of these models to define the role of inflammation, skeletal cell lineages, signaling pathways, the extracellular matrix, osteoclasts and angiogenesis. These genetic mouse models promise to change the field of orthopedic surgery to help establish genetic predispositions for delayed repair, develop models of non-union that mimic the human conditions and elaborate new therapeutic approaches to enhance bone regeneration.
Collapse
Affiliation(s)
- Rana Abou-Khalil
- INSERM UMR1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Céline Colnot
- INSERM UMR1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France.
| |
Collapse
|