301
|
Vascular endothelial growth factor: an essential component of angiogenesis and fracture healing. HSS J 2010; 6:85-94. [PMID: 19763695 PMCID: PMC2821499 DOI: 10.1007/s11420-009-9129-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 07/14/2009] [Indexed: 02/07/2023]
Abstract
Fractures require adequate stability and blood supply to heal. The vascular supply to long bones is compromised in a fracture, and the ability to heal hinges on the ability of new blood vessels to proliferate from surrounding vessels in a process known as angiogenesis. This process is largely driven by the growth factor, vascular endothelial growth factor (VEGF), whose levels are increased locally and systemically during fracture healing. VEGF is involved in many steps throughout the fracture healing cascade, from initially being concentrated in fracture hematoma, to the promotion of bone turnover during the final remodeling phase. This article reviews the current literature surrounding the role of VEGF and other growth factors in reestablishing vascular supply to fractured bone, as well as medications and surgical techniques that may inhibit this process.
Collapse
|
302
|
Anastasilakis AD, Toulis KA, Polyzos SA, Terpos E. RANKL inhibition for the management of patients with benign metabolic bone disorders. Expert Opin Investig Drugs 2010; 18:1085-102. [PMID: 19558335 DOI: 10.1517/13543780903048929] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The receptor activator of NF-kappaB ligand (RANKL) is a member of the TNF receptor superfamily, essential for osteoclastogenesis. It binds to its receptor activator of NF-kappaB on the surface of osteoclast precursors and enhances their differentiation, survival and fusion, while it activates mature osteoclasts and inhibits their apoptosis. The effects of RANKL are counteracted by osteoprotegerin (OPG), a neutralizing decoy receptor. Derangement of the balance in RANKL/OPG action is implicated in the pathophysiology of metabolic bone diseases, including osteoporosis. Current therapies used to prevent or treat metabolic bone diseases are thought to act, at least in part, through modification of the RANKL/OPG dipole. The idea of using a molecule that could specifically bind and neutralize RANKL to decrease bone resorption and subsequent bone loss is appealing. Recombinant OPG was initially tested. Denosumab, a fully human monoclonal antibody against RANKL, is a promising antiresorptive agent under investigation. It rapidly decreases bone turnover markers resulting in a significant increase in bone mineral density and reduction in fracture risk. However, because receptor activator of NF-kappaB activation by RANKL is also essential for T-cell growth and dendritic-cell function, inhibition of its action could simultaneously affect the immune system, leading to susceptibility in infections or malignancies.
Collapse
|
303
|
Deschaseaux F, Pontikoglou C, Sensébé L. Bone regeneration: the stem/progenitor cells point of view. J Cell Mol Med 2010; 14:103-15. [PMID: 19840188 PMCID: PMC3837599 DOI: 10.1111/j.1582-4934.2009.00878.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 08/04/2009] [Indexed: 02/06/2023] Open
Abstract
After bone injuries, several molecular mechanisms establish bone repair from stem/progenitor cells. Inflammation factors attract regenerative cells which expand and differentiate in order to build up a bone highly similar to that before injury. Bone marrow (BM) mesenchymal stem cells (MSCs) as skeletal stem cells and endothelial progenitors (EPCs) are at the origin of such reparation mechanisms. However, discrepancies exist about their identities. Although cultured MSCs are extensively described, their in vivo native forms are poorly known. In addition, recent experiments show that several types of EPC exist. We therefore review up-to-date data on the characterization of such stem/progenitor cells and propose a new point of view of their function in bone regeneration.
Collapse
Affiliation(s)
- Frédéric Deschaseaux
- Etablissement Français du Sang Centre-Atlantique, Groupe de Recherche sur les Cellules Souches Mésenchymateuses (GECSoM), Tours, France.
| | | | | |
Collapse
|
304
|
Lienau J, Schmidt-Bleek K, Peters A, Weber H, Bail HJ, Duda GN, Perka C, Schell H. Insight into the Molecular Pathophysiology of Delayed Bone Healing in a Sheep Model. Tissue Eng Part A 2010; 16:191-9. [DOI: 10.1089/ten.tea.2009.0187] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jasmin Lienau
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Peters
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Hauke Weber
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Hermann J. Bail
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Perka
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Hanna Schell
- Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
305
|
Coulibaly MO, Sietsema DL, Burgers TA, Mason J, Williams BO, Jones CB. Recent advances in the use of serological bone formation markers to monitor callus development and fracture healing. Crit Rev Eukaryot Gene Expr 2010; 20:105-27. [PMID: 21133841 PMCID: PMC3070362 DOI: 10.1615/critreveukargeneexpr.v20.i2.20] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The failure of an osseous fracture to heal, or the development of a nonunion, is common; however, current diagnostic measures lack the capability of early and reliable detection of such events. Analyses of radiographic imaging and clinical examination, in combination, remain the gold standard for diagnosis; however, these methods are not reliable for early detection. Delayed diagnosis of a nonunion is costly from both the patient and treatment standpoints. In response, repeated efforts have been made to identify bone metabolic markers as diagnostic or prognostic tools for monitoring bone healing. Thus far, the evidence regarding a correlation between the kinetics of most bone metabolic markers and nonunion is very limited. With the aim of classifying the role of biological pathways of bone metabolism and of understanding bone conditions in the development of osteoporosis, advances have been made in our knowledge of the molecular basis of bone remodeling, fracture healing, and its failure. Procollagen type I amino-terminal propeptide has been shown to be a reliable bone formation marker in osteoporosis therapy and its kinetics during fracture healing has been recently described. In this article, we suggest that procollagen type I amino-terminal propeptide presents a good opportunity for early detection of nonunion. We also review the role and potential of serum PINP, as well as other markers, as indications of fracture healing.
Collapse
Affiliation(s)
- Marlon O. Coulibaly
- Grand Rapids Medical Education and Research Center, Grand Rapids, Michigan, USA
| | - Debra L. Sietsema
- Orthopaedic Associates of Michigan, Grand Rapids, Michigan, USA
- Center for Skeletal Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
- Michigan State University, Grand Rapids, Michigan, USA
| | - Travis A. Burgers
- Center for Skeletal Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jim Mason
- Center for Skeletal Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Bart O. Williams
- Center for Skeletal Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Clifford B. Jones
- Orthopaedic Associates of Michigan, Grand Rapids, Michigan, USA
- Center for Skeletal Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
- Michigan State University, Grand Rapids, Michigan, USA
| |
Collapse
|
306
|
Dose effect of tumor necrosis factor-alpha on in vitro osteogenic differentiation of mesenchymal stem cells on biodegradable polymeric microfiber scaffolds. Biomaterials 2009; 31:1666-75. [PMID: 19963268 DOI: 10.1016/j.biomaterials.2009.11.058] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 11/18/2009] [Indexed: 02/08/2023]
Abstract
This study presents a first step in the development of a bone tissue engineering strategy to trigger enhanced osteogenesis by modulating inflammation. This work focused on characterizing the effects of the concentration of a pro-inflammatory cytokine, tumor necrosis factor alpha (TNF-alpha), on osteogenic differentiation of mesenchymal stem cells (MSCs) grown in a 3D culture system. MSC osteogenic differentiation is typically achieved in vitro through a combination of osteogenic supplements that include the anti-inflammatory corticosteroid dexamethasone. Although simple, the use of dexamethasone is not clinically realistic, and also hampers in vitro studies of the role of inflammatory mediators in wound healing. In this study, MSCs were pre-treated with dexamethasone to induce osteogenic differentiation, and then cultured in biodegradable electrospun poly(epsilon-caprolactone) (PCL) scaffolds, which supported continued MSC osteogenic differentiation in the absence of dexamethasone. Continuous delivery of 0.1 ng/mL of recombinant rat TNF-alpha suppressed osteogenic differentiation of rat MSCs over 16 days, which was likely the result of residual dexamethasone antagonizing TNF-alpha signaling. Continuous delivery of a higher dose, 5 ng/mL TNF-alpha, stimulated osteogenic differentiation for a few days, and 50 ng/mL TNF-alpha resulted in significant mineralized matrix deposition over the course of the study. These findings suggest that the pro-inflammatory cytokine TNF-alpha stimulates osteogenic differentiation of MSCs, an effect that can be blocked by the presence of anti-inflammatory agents like dexamethasone, with significant implications on the interplay between inflammation and tissue regeneration.
Collapse
|
307
|
Wittrant Y, Gorin Y, Mohan S, Wagner B, Abboud-Werner SL. Colony-stimulating factor-1 (CSF-1) directly inhibits receptor activator of nuclear factor-{kappa}B ligand (RANKL) expression by osteoblasts. Endocrinology 2009; 150:4977-88. [PMID: 19819976 PMCID: PMC2775986 DOI: 10.1210/en.2009-0248] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Colony-stimulating factor-1 (CSF-1), released by osteoblasts, stimulates the proliferation of osteoclast progenitors via the c-fms receptor (CSF-1R) and, in combination with receptor activator of nuclear factor-kappaB ligand (RANKL), leads to the formation of mature osteoclasts. Whether the CSF-1R is expressed by osteoblasts and mediates specific biological effects in osteoblasts has not been explored. Wild-type primary calvaria osteoblasts (OB) were analyzed for CSF-1R expression (RT-PCR and Western blot) and functionality (immunocomplex kinase assay). OB were serum starved for 24 h, and the effect of CSF-1 (0-100 ng/ml) on OB biological activities was determined at 48 h. In wild-type mouse bone marrow cultures, CSF-1 was tested for its effect on RANKL mRNA and osteoclast formation. Because ROS influence osteoblast RANKL expression, studies analyzed the effect of CSF-1 on reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity and Nox1 and Nox4 proteins. Results indicate that OB express CSF-1R mRNA and protein and that CSF-1R could be phosphorylated in the presence of CSF-1. In osteoblasts, CSF-1 decreased RANKL mRNA in a dose- and time-dependent manner. Incubation of bone marrow cultures with CSF-1 resulted in a significant decline in tartrate-resistant acid phosphatase (TRACP) activity and CTR expression. RANKL-decreased expression by CSF-1 was correlated with a decrease of NADPH oxidase activity as well as Nox1 and Nox4 protein levels. These findings provide the first evidence that osteoblasts express CSF-1R and are a target for CSF-1 ligand. CSF-1-mediated inhibition of RANKL expression on osteoblasts may provide an important mechanism for coupling bone formation/resorption and preventing excessive osteoclastogenesis during normal skeletal growth.
Collapse
Affiliation(s)
- Y Wittrant
- South Texas Veteran's Health Care System, Audi L. Murphy Division, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, Texas 78229, USA
| | | | | | | | | |
Collapse
|
308
|
Ota N, Takaishi H, Kosaki N, Takito J, Yoda M, Tohmonda T, Kimura T, Okada Y, Yasuda H, Kawaguchi H, Matsumoto M, Chiba K, Ikegami H, Toyama Y. Accelerated cartilage resorption by chondroclasts during bone fracture healing in osteoprotegerin-deficient mice. Endocrinology 2009; 150:4823-34. [PMID: 19819969 DOI: 10.1210/en.2009-0452] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG), a decoy receptor of RANKL, maintain bone mass by regulating the differentiation of osteoclasts, which are bone-resorbing cells. Endochondral bone ossification and bone fracture healing involve cartilage resorption, a less well-understood process that is needed for replacement of cartilage by bone. Here we describe the role of OPG produced by chondrocytes in chondroclastogenesis. Fracture healing in OPG(-/-) mice showed faster union of the fractured bone, faster resorption of the cartilaginous callus, and an increased number of chondroclasts at the chondroosseous junctions compared with that in wild-type littermates. When a cultured pellet of OPG(-/-) chondrocytes was transplanted beneath the kidney capsule, the pellet recruited many chondroclasts. The pellet showed the ability to induce tartrate-resistant acid phosphatase-positive multinucleated cells from RAW 264.7 cells in vitro. Finally, OPG(-/-) chondrocytes (but not wild-type chondrocytes) cultured with spleen cells induced many tartrate-resistant acid phosphatase-positive multinucleated cells. The expression of RANKL and OPG in chondrocytes was regulated by several osteotropic factors including 1,25-dihydroxyvitamin D(3), PTHrP, IL-1alpha, and TNF-alpha. Thus, local OPG produced by chondrocytes probably controls cartilage resorption as a negative regulator for chondrocyte-dependent chondroclastogenesis.
Collapse
Affiliation(s)
- Norikazu Ota
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
309
|
Cenni E, Ciapetti G, Granchi D, Fotia C, Perut F, Giunti A, Baldini N. Endothelial cells incubated with platelet-rich plasma express PDGF-B and ICAM-1 and induce bone marrow stromal cell migration. J Orthop Res 2009; 27:1493-8. [PMID: 19396860 DOI: 10.1002/jor.20896] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Platelet-rich plasma (PRP) is used to accelerate bone repair through the growth factors released by platelets. The purpose of this study was to evaluate if PRP induce human umbilical vein endothelial cells (HUVEC) to express mRNA for osteogenic growth factors and stimulate the migration of bone marrow stromal cell (BMSC). The effects of PRP were compared to those induced by vascular endothelial growth factor-A (VEGF-A) or, as a negative control, by platelet poor plasma (PPP). After incubation with PRP, but not with PPP, HUVEC showed an increased expression of mRNA for platelet derived growth factor-B (PDGF-B), and this effect was not inhibited by an anti-VEGF-A antibody. The migration of BMSC was more stimulated by HUVEC incubated with PRP than by HUVEC incubated with low serum medium or PPP. Besides, PRP increased the expression of intercellular adhesion molecule-1 (ICAM-1) and osteoprotegerin, but did not affect the expression either of the receptor activator for nuclear factor kappaB ligand (RANKL) or of RANK. These findings support the hypothesis that PRP contribute to bone repair by favoring the pro-osteogenic function of endothelial cells, including the recruitment of osteoblast precursors and the expression of adhesion molecules for monocyte/macrophages, while inhibiting their pro-osteolytic properties.
Collapse
Affiliation(s)
- Elisabetta Cenni
- Laboratory for Pathophysiology of Orthopaedic Implants, Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
310
|
Alblowi J, Kayal RA, Siqueira M, Siqueria M, McKenzie E, Krothapalli N, McLean J, Conn J, Nikolajczyk B, Einhorn TA, Gerstenfeld L, Graves DT. High levels of tumor necrosis factor-alpha contribute to accelerated loss of cartilage in diabetic fracture healing. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1574-85. [PMID: 19745063 DOI: 10.2353/ajpath.2009.090148] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Diabetes interferes with fracture repair; therefore, we investigated mechanisms of impaired fracture healing in a model of multiple low-dose streptozotocin-induced diabetes. Microarray and gene set enrichment analysis revealed an up-regulation of gene sets related to inflammation, including tumor necrosis factor (TNF) signaling in the diabetic group, when cartilage is being replaced by bone on day 16, but not on days 12 or 22. This change coincided with elevated osteoclast numbers and accelerated removal of cartilage in the diabetic group (P < 0.05), which was reflected by smaller callus size. When diabetic mice were treated with the TNF-specific inhibitor, pegsunercept, the number of osteoclasts, cartilage loss, and number of TNF-alpha and receptor activator for nuclear factor kB ligand positive chondrocytes were significantly reduced (P < 0.05). The transcription factor forkhead box 01 (FOXO1) was tested for mediating TNF stimulation of osteoclastogenic and inflammatory factors in bone morphogenetic protein 2 pretreated ATDC5 and C3H10T1/2 chondrogenic cells. FOXO1 knockdown by small-interfering RNA significantly reduced TNF-alpha, receptor activator for nuclear factor kB ligand, macrophage colony-stimulating factor, interleukin-1alpha, and interleukin-6 mRNA compared with scrambled small-interfering RNA. An association between FOXO1 and the TNF-alpha promoter was demonstrated by chromatin immunoprecipitation assay. Moreover, diabetes increased FOXO1 nuclear translocation in chondrocytes in vivo and increased FOXO1 DNA binding activity in diabetic fracture calluses (P < 0.05). These results suggest that diabetes-enhanced TNF-alpha increases the expression of resorptive factors in chondrocytes through a process that involves activation of FOXO1 and that TNF-alpha dysregulation leads to enhanced osteoclast formation and accelerated loss of cartilage.
Collapse
Affiliation(s)
- Jazia Alblowi
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
311
|
Mountziaris PM, Mikos AG. Modulation of the inflammatory response for enhanced bone tissue regeneration. TISSUE ENGINEERING PART B-REVIEWS 2009; 14:179-86. [PMID: 18544015 DOI: 10.1089/ten.teb.2008.0038] [Citation(s) in RCA: 342] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Proinflammatory cytokines are infamous for their catabolic effects on tissues and joints in both inflammatory diseases and following the implantation of biomedical devices. However, recent studies indicate that many of these same molecules are critical for triggering tissue regeneration following injury. This review will discuss the role of inflammatory signals in regulating bone regeneration and the impact of both immunomodulatory and antiinflammatory pharmacologic agents on fracture healing, to demonstrate the importance of incorporating rational control of inflammation into the design of tissue engineering strategies.
Collapse
|
312
|
Osteogenic Differentiation is Selectively Promoted by Morphogenetic Signals from Chondrocytes and Synergized by a Nutrient Rich Growth Environment. Connect Tissue Res 2009. [DOI: 10.1080/03008200390152142] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2023]
|
313
|
Sundelacruz S, Kaplan DL. Stem cell- and scaffold-based tissue engineering approaches to osteochondral regenerative medicine. Semin Cell Dev Biol 2009; 20:646-55. [PMID: 19508851 PMCID: PMC2737137 DOI: 10.1016/j.semcdb.2009.03.017] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 03/30/2009] [Indexed: 11/24/2022]
Abstract
In osteochondral tissue engineering, cell recruitment, proliferation, differentiation, and patterning are critical for forming biologically and structurally viable constructs for repair of damaged or diseased tissue. However, since constructs prepared ex vivo lack the multitude of cues present in the in vivo microenvironment, cells often need to be supplied with external biological and physical stimuli to coax them toward targeted tissue functions. To determine which stimuli to present to cells, bioengineering strategies can benefit significantly from endogenous examples of skeletogenesis. As an example of developmental skeletogenesis, the developing limb bud serves as an excellent model system in which to study how osteochondral structures form from undifferentiated precursor cells. Alongside skeletal formation during embryogenesis, bone also possesses innate regenerative capacity, displaying remarkable ability to heal after damage. Bone fracture healing shares many features with bone development, driving the hypothesis that the regenerative process generally recapitulates development. Similarities and differences between the two modes of bone formation may offer insight into the special requirements for healing damaged or diseased bone. Thus, endogenous fracture healing, as an example of regenerative skeletogenesis, may also inform bioengineering strategies. In this review, we summarize the key cellular events involving stem and progenitor cells in developmental and regenerative skeletogenesis, and discuss in parallel the corresponding cell- and scaffold-based strategies that tissue engineers employ to recapitulate these events in vitro.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| |
Collapse
|
314
|
Tosounidis T, Kontakis G, Nikolaou V, Papathanassopoulos A, Giannoudis PV. Fracture healing and bone repair: an update. TRAUMA-ENGLAND 2009; 11:145-156. [DOI: 10.1177/1460408609335922] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Bone healing represents a physiological process of repair and restoration of function. Recent advances in a variety of medical disciplines have enabled scientists and clinicians to characterise this phenomenon at the molecular level. A number of molecular mediators and cells interact utilising different pathways. Despite the involvement of many local and systemic factors failure of the naturally occurring mechanisms can occur leading to either delayed union or non-union. This review article is focused on the recent understanding of the mechanisms governing the bone repair process.
Collapse
Affiliation(s)
| | - George Kontakis
- Department of Orthopaedics and Traumatology, University of Crete, Greece
| | - Vassilis Nikolaou
- Academic Department of Trauma and Orthopaedics, University of Leeds, UK
| | | | - Peter V Giannoudis
- Academic Department of Trauma and Orthopaedics, University of Leeds, UK,
| |
Collapse
|
315
|
Gaur T, Wixted JJ, Hussain S, O’Connell S, Morgan EF, Ayers D, Komm BS, Bodine PV, Stein GS, Lian JB. Secreted frizzled related protein 1 is a target to improve fracture healing. J Cell Physiol 2009; 220:174-81. [PMID: 19301255 PMCID: PMC2756719 DOI: 10.1002/jcp.21747] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Genetic studies have identified a high bone mass of phenotype in both human and mouse when canonical Wnt signaling is increased. Secreted frizzled related protein 1 (sFRP1) is one of several Wnt antagonists and among the loss-of-function mouse models in which 32-week-old mice exhibit a high bone mass phenotype. Here we show that impact fracture healing is enhanced in this mouse model of increased Wnt signaling at a physiologic level in young (8 weeks) sFRP1(-/-) mice which do not yet exhibit significant increases in BMD. In vivo deletion of sFRP1 function improves fracture repair by promoting early bone union without adverse effects on the quality of bone tissue reflected by increased mechanical strength. We observe a dramatic reduction of the cartilage callous, increased intramembranous bone formation with bone bridging by 14 days, and early bone remodeling during the 28-day fracture repair process in the sFRP1(-/-) mice. Our molecular analyses of gene markers indicate that the effect of sFRP1 loss-of-function during fracture repair is to accelerate bone healing after formation of the initial hematoma by directing mesenchymal stem cells into the osteoblast lineage via the canonical pathway. Further evidence to support this conclusion is the observation of maximal sFRP1 levels in the cartilaginous callus of a WT mouse. Hence sFRP1(-/-) mouse progenitor cells are shifted directly into the osteoblast lineage. Thus, developing an antagonist to specifically inhibit sFRP1 represents a safe target for stimulating fracture repair and bone formation in metabolic bone disorders, osteoporosis and aging.
Collapse
Affiliation(s)
- Tripti Gaur
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, MA 01655
| | - John J. Wixted
- Department of Orthopaedic Surgery and Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655
| | - Sadiq Hussain
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, MA 01655
| | - Shannon O’Connell
- Department of Orthopaedic Surgery and Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655
| | - Elise F. Morgan
- Department of Aerospace and Mechanical Engineering, Boston University, Boston, MA 02215
| | - David Ayers
- Department of Orthopaedic Surgery and Rehabilitation, University of Massachusetts Medical School, Worcester, MA 01655
| | - Barry S. Komm
- Women’s Health and Musculoskeletal Biology, Wyeth Research, Collegeville, Pennsylvania
| | - Peter V. Bodine
- Women’s Health and Musculoskeletal Biology, Wyeth Research, Collegeville, Pennsylvania
| | - Gary S. Stein
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jane B. Lian
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, MA 01655
| |
Collapse
|
316
|
Geusens P. Emerging treatments for postmenopausal osteoporosis - focus on denosumab. Clin Interv Aging 2009; 4:241-50. [PMID: 19554095 PMCID: PMC2697589 DOI: 10.2147/cia.s3333] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Indexed: 01/23/2023] Open
Abstract
The pathway of the receptor activator of the nuclear factor κB ligand (RANKL), RANK and osteoprotegerin (OPG) plays a central role in coupling bone formation and resorption during normal bone turnover and in a wide spectrum of diseases characterized by disturbed bone remodeling, increased bone resorption and bone destruction (osteoporosis, Paget’s disease of bone, rheumatoid arthritis [RA], metastatic bone disease). Clinical trials indicate that denosumab, a RANKL-specific recombinant humanized monoclonal antibody, is effective in suppressing bone resorption, resulting in increase in bone mineral density (BMD) in post-menopausal women with low BMD, and has the potential to prevent progression of erosions in RA and of skeletal-related events in metastatic bone disease. The effects on fracture reduction in postmenopausal osteoporosis are awaited from the recently finished FREEDOM study. In clinical trials with denosumab, overall adverse events were similar to placebo or comparators, indicating a favorable safety profile in these diseases, which until now have been available up to 4 years, but data on long-term safety will be needed.
Collapse
Affiliation(s)
- Piet Geusens
- Department of Internal Medicine, Subdivision of Rheumatology, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
317
|
Colburn NT, Zaal KJM, Wang F, Tuan RS. A role for gamma/delta T cells in a mouse model of fracture healing. ARTHRITIS AND RHEUMATISM 2009; 60:1694-703. [PMID: 19479830 PMCID: PMC2697263 DOI: 10.1002/art.24520] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Fractures can initiate an immune response that disturbs osteoblastic and osteoclastic cellular homeostasis through cytokine production and release. The aim of our study was to investigate gamma/delta T cells, innate lymphocytes known to be involved in tissue repair, as potential cellular components of the osteoimmune system's response to an in vivo model of bone injury. The absence of such cells or their effector cytokines influences the fate of other responder cells in proliferation, differentiation, matrix production, and ultimate callus formation. METHODS Tibia fractures were created in 60 gamma/delta T cell-deficient mice (also called delta T cell receptor [TCR]-knockout mice) and 60 control C57BL/6 mice. Analysis included radiographs, basic histology, mechanical testing, flow cytometry, and immunohistochemical localization of gamma/delta TCR-positive subsets from control animals and of CD44 expression from both groups, as well as enzyme-linked immunosorbent assay for the effector cytokines interleukin-2 (IL-2), interferon-gamma (IFNgamma), and IL-6. RESULTS Animals deficient in gamma/delta T cells demonstrated more mature histologic elements and quantitative increases in the expression of major bone (bone sialoprotein) and cartilage (type II collagen) matrix proteins and in the expression of bone morphogenetic protein 2 at a critical reparative phase. Moreover, only gamma/delta T cell-deficient animals had a decrease in the osteoprogenitor antiproliferative cytokines IL-6 and IFNgamma at the reparative phase. The result was improved stability at the repair site and an overall superior biomechanical strength in gamma/delta T cell-deficient mice compared with controls. CONCLUSION The evidence for a role of gamma/delta T cells in the context of skeletal injury demonstrates the importance of the immune system's effect on bone biology, which is relevant to the field of osteoimmunology, and offers a potential molecular platform from which to develop essential therapeutic strategies.
Collapse
MESH Headings
- Animals
- Bone Matrix/metabolism
- Cartilage/metabolism
- Cell Differentiation/physiology
- Cell Proliferation
- Cytokines/metabolism
- Fracture Healing/physiology
- Hyaluronan Receptors/metabolism
- Immunity, Innate/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Animal
- Osteoblasts/metabolism
- Osteoblasts/pathology
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/physiology
- T-Lymphocytes/pathology
- T-Lymphocytes/physiology
Collapse
Affiliation(s)
- Nona T Colburn
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland 20892, USA.
| | | | | | | |
Collapse
|
318
|
Bais M, McLean J, Sebastiani P, Young M, Wigner N, Smith T, Kotton DN, Einhorn TA, Gerstenfeld LC. Transcriptional analysis of fracture healing and the induction of embryonic stem cell-related genes. PLoS One 2009; 4:e5393. [PMID: 19415118 PMCID: PMC2673045 DOI: 10.1371/journal.pone.0005393] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 03/25/2009] [Indexed: 11/23/2022] Open
Abstract
Fractures are among the most common human traumas. Fracture healing represents a unique temporarily definable post-natal process in which to study the complex interactions of multiple molecular events that regulate endochondral skeletal tissue formation. Because of the regenerative nature of fracture healing, it is hypothesized that large numbers of post-natal stem cells are recruited and contribute to formation of the multiple cell lineages that contribute to this process. Bayesian modeling was used to generate the temporal profiles of the transcriptome during fracture healing. The temporal relationships between ontologies that are associated with various biologic, metabolic, and regulatory pathways were identified and related to developmental processes associated with skeletogenesis, vasculogenesis, and neurogenesis. The complement of all the expressed BMPs, Wnts, FGFs, and their receptors were related to the subsets of transcription factors that were concurrently expressed during fracture healing. We further defined during fracture healing the temporal patterns of expression for 174 of the 193 genes known to be associated with human genetic skeletal disorders. In order to identify the common regulatory features that might be present in stem cells that are recruited during fracture healing to other types of stem cells, we queried the transcriptome of fracture healing against that seen in embryonic stem cells (ESCs) and mesenchymal stem cells (MSCs). Approximately 300 known genes that are preferentially expressed in ESCs and approximately 350 of the known genes that are preferentially expressed in MSCs showed induction during fracture healing. Nanog, one of the central epigenetic regulators associated with ESC stem cell maintenance, was shown to be associated in multiple forms or bone repair as well as MSC differentiation. In summary, these data present the first temporal analysis of the transcriptome of an endochondral bone formation process that takes place during fracture healing. They show that neurogenesis as well as vasculogenesis are predominant components of skeletal tissue formation and suggest common pathways are shared between post-natal stem cells and those seen in ESCs.
Collapse
Affiliation(s)
- Manish Bais
- Orthopaedic Research Laboratory, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jody McLean
- Orthopaedic Research Laboratory, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Paola Sebastiani
- School of Public Health, Boston University Medical Center, Boston, Massachusetts, United States of America
| | - Megan Young
- Orthopaedic Research Laboratory, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Nathan Wigner
- Orthopaedic Research Laboratory, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Temple Smith
- Department of Biomedical Engineering, Boston University School of Engineering, Boston, Massachusetts, United States of America
| | - Darrell N. Kotton
- Department of Medicine, Pulmonary Center Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Thomas A. Einhorn
- Orthopaedic Research Laboratory, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Louis C. Gerstenfeld
- Orthopaedic Research Laboratory, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
319
|
Lee JS, Ryu CH, Moon NH, Kim SJ, Park SY, Suh KT. Changes in serum levels of receptor activator of nuclear factor-kappaB ligand, osteoprotegerin, IL-6 and TNF-alpha in patients with a concomitant head injury and fracture. Arch Orthop Trauma Surg 2009; 129:711-8. [PMID: 18427820 DOI: 10.1007/s00402-008-0632-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Several reports indicated that interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF- alpha) play important regulatory roles in bone remodeling and homeostasis. In addition, receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG) have been shown to be important regulators of osteoclastogenesis during bone remodeling, and their expressions were examined during fracture healing in a mouse model of tibial fracture. However, studies linking RANKL, OPG, IL-6 and TNF-alpha in patients with head injury and fracture are lacking. PATIENTS AND METHODS Within the first few hours of admission to hospital and at 4, 8, and 12 weeks after the injury, we evaluated changes in serum levels of RANKL, OPG, IL-6 and TNF-alpha in 24 male patients with a concomitant head injury and fracture and in 26 male patients with fracture only. These levels were compared with those found in 36 healthy controls. RESULTS The RANKL/OPG ratios were found to significantly lower in patients with a concomitant head injury and fracture than in the controls immediately after admission and at 4, 8, and 12 weeks after the injury. In addition, RANKL/OPG ratios were significantly lower in patients with a concomitant head injury and fracture than in those with fracture at 8 and 12 weeks after the injury. The serum IL-6 levels were significantly higher in patients with a concomitant head injury and fracture than in the controls upon admission, and at 4, 8, and 12 weeks after the injury. Moreover, the serum IL-6 levels were significantly higher in patients with a head injury and fracture than in those with just a fracture at 4, 8, and 12 weeks after the injury. CONCLUSIONS Based on these changes in the profiles of RANKL, OPG, and IL-6 and the RANKL/OPG ratio, altered repair of a fracture can occur in patients with a concomitant head injury and fracture.
Collapse
Affiliation(s)
- Jung Sub Lee
- Department of Orthopaedic Surgery, Medical Research Institute, Pusan National University School of Medicine, Pusan, South Korea
| | | | | | | | | | | |
Collapse
|
320
|
Kayal RA, Alblowi J, McKenzie E, Krothapalli N, Silkman L, Gerstenfeld L, Einhorn TA, Graves DT. Diabetes causes the accelerated loss of cartilage during fracture repair which is reversed by insulin treatment. Bone 2009; 44:357-63. [PMID: 19010456 PMCID: PMC2700945 DOI: 10.1016/j.bone.2008.10.042] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 10/01/2008] [Accepted: 10/14/2008] [Indexed: 11/15/2022]
Abstract
Fracture healing in diabetic individuals and in animal models of diabetes is impaired. To investigate mechanisms by which diabetes may affect fracture healing we focused on the transition from cartilage to bone, a midpoint in the fracture healing process. Femoral fractures were induced in mice rendered diabetic by multiple low dose streptozotocin treatment and compared to matching normoglycemic mice. One group of diabetic animals was treated with slow release insulin to maintain normal serum glucose levels. The results indicate that there was relatively little difference in the initial formation of the fracture callus on day 10. However, on day 16 the diabetic group had significantly smaller callus, greater loss of cartilage and enhanced osteoclastogenesis that was normalized by treatment with insulin when assessed by histomorphometric analysis. Chondrocyte apoptosis was significantly higher in diabetic mice and this increase was blocked by insulin. These changes were accompanied by diabetes-increased mRNA levels of RANKL, TNF-alpha, and ADAMTS-4 and -5 measured by real-time PCR, which was reversed by insulin treatment. On days 16 and 22 bone formation within the callus of diabetic mice was significantly less than the normoglycemic and brought to normal levels by insulin treatment. These results suggest that a significant effect of diabetes on fracture healing is increased chondrocyte apoptosis and osteoclastogenesis that accelerates the loss of cartilage and reduces the anlage for endochondral bone formation during fracture repair. That insulin reverses these effects demonstrates that they are directly related to the diabetic condition.
Collapse
Affiliation(s)
- Rayyan A. Kayal
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA
| | - Jazia Alblowi
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA
| | - Erin McKenzie
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA
| | - Nanarao Krothapalli
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA
| | - Lee Silkman
- Department of Orthopedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Louis Gerstenfeld
- Department of Orthopedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Thomas A. Einhorn
- Department of Orthopedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Dana T. Graves
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA
| |
Collapse
|
321
|
Gerstenfeld LC, Sacks DJ, Pelis M, Mason ZD, Graves DT, Barrero M, Ominsky MS, Kostenuik PJ, Morgan EF, Einhorn TA. Comparison of effects of the bisphosphonate alendronate versus the RANKL inhibitor denosumab on murine fracture healing. J Bone Miner Res 2009; 24:196-208. [PMID: 19016594 PMCID: PMC6961532 DOI: 10.1359/jbmr.081113] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 08/25/2008] [Accepted: 10/28/2008] [Indexed: 01/21/2023]
Abstract
The role of osteoclast-mediated resorption during fracture healing was assessed. The impact of two osteoclast inhibitors with different mechanisms of action, alendronate (ALN) and denosumab (DMAB), were examined during fracture healing. Male human RANKL knock-in mice that express a chimeric (human/murine) form of RANKL received unilateral transverse femur fractures. Mice were treated biweekly with ALN 0.1 mg/kg, DMAB 10 mg/kg, or PBS (control) 0.1 ml until death at 21 and 42 days after fracture. Treatment efficacy assessed by serum levels of TRACP 5b showed almost a complete elimination of TRACP 5b levels in the DMAB-treated animals but only approximately 25% reduction of serum levels in the ALN-treated mice. Mechanical testing showed that fractured femurs from both ALN and DMAB groups had significantly increased mechanical properties at day 42 compared with controls. muCT analysis showed that callus tissues from DMAB-treated mice had significantly greater percent bone volume and BMD than did both control and ALN-treated tissues at both 21 and 42 days, whereas ALN-treated bones only had greater percent bone volume and BMC than control at 42 days. Qualitative histological analysis showed that the 21-and 42-day ALN and DMAB groups had greater amounts of unresorbed cartilage or mineralized cartilage matrix compared with the controls, whereas unresorbed cartilage could still be seen in the DMAB groups at 42 days after fracture. Although ALN and DMAB delayed the removal of cartilage and the remodeling of the fracture callus, this did not diminish the mechanical integrity of the healing fractures in mice receiving these treatments. In contrast, strength and stiffness were enhanced in these treatment groups compared with control bones.
Collapse
Affiliation(s)
- Louis C Gerstenfeld
- Orthopaedic Research Laboratory, Department of Orthopedic Surgery, Boston University School of Medicine, Boston, Massachusetts 02118, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
McCarthy HS, Williams JH, Davie MW, Marshall MJ. Platelet-derived growth factor stimulates osteoprotegerin production in osteoblastic cells. J Cell Physiol 2009; 218:350-4. [DOI: 10.1002/jcp.21600] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
323
|
Goodman ML, Chen S, Yang FC, Chan RJ. Novel method of murine embryonic stem cell-derived osteoclast development. Stem Cells Dev 2009; 18:195-200. [PMID: 18616393 PMCID: PMC2975428 DOI: 10.1089/scd.2008.0193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2008] [Accepted: 07/10/2008] [Indexed: 10/21/2022] Open
Abstract
Murine embryonic stem (mES) cells are self-renewing pluripotent cells that bear the capacity to differentiate into ectoderm-, endoderm-, and mesoderm-derived tissues. In suspension culture, embryonic stem (ES) cells grow into spherical embryoid bodies (EBs) and are useful for the study of specific gene products in the development and function of various tissue types. Osteoclasts are hematopoietic stem cell-derived cells that participate in bone turnover by secreting resorptive molecules such as hydrochloric acid and acidic proteases, which degrade the bone extracellular matrix. Aberrant osteoclast function leads to dysplastic, erosive, and sclerosing bone diseases. Previous studies have reported the derivation of osteoclasts from mES cells; however, most of these protocols require coculture with stromal cell lines. We describe two simplified, novel methods of stromal cell-independent ES cell-derived osteoclast development.
Collapse
Affiliation(s)
- Michael L. Goodman
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Shi Chen
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Feng-Chun Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Rebecca J. Chan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Medical and Molecular Genetics, the Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
324
|
Sarahrudi K, Mousavi M, Grossschmidt K, Sela N, König F, Vécsei V, Aharinejad S. The impact of colony-stimulating factor-1 on fracture healing: an experimental study. J Orthop Res 2009; 27:36-41. [PMID: 18634005 DOI: 10.1002/jor.20680] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The role of colony stimulating factor-1 (CSF-1) in the regulation of osteoclasts and bone remodeling suggests that CSF-1 may also be involved in regulation of bone healing. The ability of CSF-1 to promote healing of bone defects was tested in a rabbit model. Twenty-four New Zeeland rabbits were included in the study. Animals were assigned to two groups: the control group (n = 12) was treated by plate fixation. The animals in the second group (n = 12) were also stabilized by conventional plating and received additionally CSF-1 for 2 weeks systemically. Histologic, histomorphometric, and radiologic examinations were performed to evaluate the healing process at 4, 8, and 12 weeks following surgery. Animals that were treated by CSF-1 produced a significantly higher amount of mineralized bone over the first 8 weeks after fracture compared to the control animals. Furthermore, a higher number of osteoclasts was found in CSF-1-treated animals within the first 8 weeks, compared to the controls. The present data emphasize for the first time the importance of CSF-1 in the bone healing. The use of CSF-1 in addition to conventional fixation might be a novel approach for the treatment of bone defects.
Collapse
Affiliation(s)
- Kambiz Sarahrudi
- Department of Traumatology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
325
|
Morgan EF, Mason ZD, Bishop G, Davis AD, Wigner NA, Gerstenfeld LC, Einhorn TA. Combined effects of recombinant human BMP-7 (rhBMP-7) and parathyroid hormone (1-34) in metaphyseal bone healing. Bone 2008; 43:1031-8. [PMID: 18761113 DOI: 10.1016/j.bone.2008.07.251] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 06/27/2008] [Accepted: 07/20/2008] [Indexed: 11/25/2022]
Abstract
Fracture healing involves multiple stages of repair and coordinated actions of multiple cell types. Consequently, it may be possible to enhance healing through treatment strategies that target more than one repair process or cell type. The goal of this study was to determine the combined effects of recombinant human bone morphogenetic protein 7 (rhBMP-7) and parathyroid hormone (PTH(1-34)) on metaphyseal bone healing. A wedge-shaped defect was created in the lateral aspect of the distal tibia in female New Zealand white rabbits (n=64) and was filled with tricalcium phosphate (TCP). Animals were assigned to four groups: 1) BMP-7 and PTH; 2) BMP-7; 3) PTH; and 4) control (TCP alone). In groups 1 and 2, 200 microg rhBMP-7 was incorporated into the TCP. Animals received daily subcutaneous injections of 10 microg/kg PTH(1-34) (groups 1 and 3) or saline (groups 2 and 4). Healing at 4 weeks was assessed using micro-computed tomography, histology, immunohistochemistry, and mechanical testing. Combined treatment with rhBMP-7 and PTH resulted in increased callus total volume (TV), mineralized volume (BV), average cross-sectional area, and bone mineral content (BMC) as compared to the control group (p<0.02). BV and BMC were also higher in the combined treatment group as compared to the BMP-7 group (p<0.02); however, tissue mineral density was highest in the BMP-7 group (p=0.002). New bone formation in the BMP-7 group was largely restricted to the defect site, while PTH promoted bone formation throughout the defect and surrounding regions. Combined treatment led to greater quantities of woven trabecular bone, increased trabecular thickness, decreased trabecular separation (p<0.04), and a trend towards increased numbers of osteoclasts (p=0.09). Combined treatment also resulted in increased torsional rigidity and compressive strength as compared to the control and BMP-7 groups (p<0.001). These results suggest that the improvements in mechanical function obtained with the combined treatment resulted from differing biological activities of rhBMP-7 and PTH. While the activities of rhBMP-7 appeared to be strictly anabolic, those of PTH appeared to work in the context of coupled remodeling. The combination of both agents led to greater bone volume as well as better microstructural organization and integration of this bone with the surrounding tissues.
Collapse
Affiliation(s)
- Elise F Morgan
- Orthopaedic and Developmental Biomechanics Laboratory, Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
326
|
Miki C, Kusunoki M, Inoue Y, Uchida K, Mohri Y, Buckels JAC, McMaster P. Remodeling of the immunoinflammatory network system in elderly cancer patients: Implications of inflamm-aging and tumor-specific hyperinflammation. Surg Today 2008; 38:873-8. [DOI: 10.1007/s00595-008-3766-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 02/01/2008] [Indexed: 10/21/2022]
|
327
|
Richardson J, Hill AM, Johnston CJC, McGregor A, Norrish AR, Eastwood D, Lavy CBD. Fracture healing in HIV-positive populations. ACTA ACUST UNITED AC 2008; 90:988-94. [DOI: 10.1302/0301-620x.90b8.20861] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Highly active anti-retroviral therapy has transformed HIV into a chronic disease with a long-term asymptomatic phase. As a result, emphasis is shifting to other effects of the virus, aside from immunosuppression and mortality. We have reviewed the current evidence for an association between HIV infection and poor fracture healing. The increased prevalence of osteoporosis and fragility fractures in HIV patients is well recognised. The suggestion that this may be purely as a result of highly active anti-retroviral therapy has been largely rejected. Apart from directly impeding cellular function in bone remodelling, HIV infection is known to cause derangement in the levels of those cytokines involved in fracture healing (particularly tumour necrosis factor-α) and appears to impair the blood supply of bone. Many other factors complicate this issue, including a reduced body mass index, suboptimal nutrition, the effects of anti-retroviral drugs and the avoidance of operative intervention because of high rates of wound infection. However, there are sound molecular and biochemical hypotheses for a direct relationship between HIV infection and impaired fracture healing, and the rewards for further knowledge in this area are extensive in terms of optimised fracture management, reduced patient morbidity and educated resource allocation. Further investigation in this area is overdue.
Collapse
Affiliation(s)
- J. Richardson
- Faculty of Medicine, Imperial College London, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK
| | - A. M. Hill
- Faculty of Medicine, Imperial College London, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK
| | - C. J. C. Johnston
- Faculty of Medicine, Imperial College London, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK
| | - A. McGregor
- Faculty of Medicine, Imperial College London, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK
| | - A. R. Norrish
- Tropical Surgery Research and Training Unit, Beit CURE Hospital, P. O. Box 36391, Lusaka, Zambia
| | - D. Eastwood
- Department of Orthopaedics, Great Ormond Street Hospital, Great Ormond Street, London WC1N 3JH, UK
| | - C. B. D. Lavy
- Nuffield Department of Orthopaedic Surgery, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford 0X3 7LD, UK
| |
Collapse
|
328
|
Jepsen KJ, Price C, Silkman LJ, Nicholls FH, Nasser P, Hu B, Hadi N, Alapatt M, Stapleton SN, Kakar S, Einhorn TA, Gerstenfeld LC. Genetic variation in the patterns of skeletal progenitor cell differentiation and progression during endochondral bone formation affects the rate of fracture healing. J Bone Miner Res 2008; 23:1204-16. [PMID: 18348700 PMCID: PMC2650253 DOI: 10.1359/jbmr.080317] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Revised: 02/20/2008] [Accepted: 03/12/2008] [Indexed: 12/30/2022]
Abstract
These studies examined how genetic differences that regulate architectural and bone material properties would be expressed during fracture healing and determine whether any of these features would affect rates of healing as defined by regain of strength. Controlled fractures were generated in three inbred strains of mice: A/J, C57Bl/6J (B6), and C3H/HeJ (C3H). Both the A/J and B6 strains showed faster healing than the C3H strain based on regains in strength and stiffness. Strain-specific architectural features such as moment of inertia, cross-sectional area, and cortical thickness were all recapitulated during the development of the callus tissues. None of these traits were directly relatable to rates of fracture healing. However, rates of healing were related to variations in the temporal patterns of chondrogenic and osteogenic lineage development. The B6 strain expressed the highest percentage of cartilage gene products and had the longest period of chondrocyte maturation and hypertrophy. The slowest healing strain (C3H) had the shortest period of chondrogenic development and earliest initiation of osteogenic development. Although the A/J strain showed an almost identical pattern of chondrogenic development as the C3H strain, A/J initiated osteogenic development several days later than C3H during fracture healing. Long bone growth plates at 28 days after birth showed similar strain-specific variation in cartilage tissue development as seen in fracture healing. Thus, the B6 strain had the largest growth plate heights, cell numbers per column, and the largest cell size, whereas the C3H columns were the shortest, had the smallest number of cells per column, and showed the smallest cell sizes. These results show that (1) different strains of mice express variations of skeletal stem cell lineage differentiation and (2) that these variations affect the rate of fracture healing.
Collapse
Affiliation(s)
- Karl J Jepsen
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, USA
| | - Christopher Price
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, USA
| | - Lee J Silkman
- Orthopaedic Research Laboratory, Boston University Medical Center, Boston, Massachusetts, USA
| | - Fred H Nicholls
- Orthopaedic Research Laboratory, Boston University Medical Center, Boston, Massachusetts, USA
| | - Phillip Nasser
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, USA
| | - Bin Hu
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, USA
| | - Nicole Hadi
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, USA
| | - Michael Alapatt
- Orthopaedic Research Laboratory, Boston University Medical Center, Boston, Massachusetts, USA
| | - Stephanie N Stapleton
- Orthopaedic Research Laboratory, Boston University Medical Center, Boston, Massachusetts, USA
| | - Sanjeev Kakar
- Orthopaedic Research Laboratory, Boston University Medical Center, Boston, Massachusetts, USA
| | - Thomas A Einhorn
- Orthopaedic Research Laboratory, Boston University Medical Center, Boston, Massachusetts, USA
| | - Louis C Gerstenfeld
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
329
|
Hallman M, Thor A. Bone substitutes and growth factors as an alternative/complement to autogenous bone for grafting in implant dentistry. Periodontol 2000 2008; 47:172-92. [PMID: 18412581 DOI: 10.1111/j.1600-0757.2008.00251.x] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
330
|
La Fontaine J, Harkless LB, Sylvia VL, Carnes D, Heim-Hall J, Jude E. Levels of endothelial nitric oxide synthase and calcitonin gene-related peptide in the Charcot foot: a pilot study. J Foot Ankle Surg 2008; 47:424-9. [PMID: 18725122 DOI: 10.1053/j.jfas.2008.05.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Indexed: 02/03/2023]
Abstract
UNLABELLED The pathogenesis of Charcot neuroarthropathy is unclear. To investigate the possibility that decreased levels of calcitonin gene-related peptide and endothelial nitric oxide synthase are involved in the process, we studied bone samples from healthy subjects (n = 4), subjects with diabetic neuropathy (n = 4), and subjects with Charcot neuroarthropathy (n = 4). A statistically significant difference was found in endothelial nitric oxide synthase expression between bone specimens in patients with diabetic neuropathy, Charcot neuroarthropathy, and normal bone (P = .008). A trend toward calcitonin gene-related peptide intensification was observed in normal bone as compared to diabetic neuropathy and Charcot neuroarthropathy bone specimens, but it did not reached statistical significance (P = .23). This pilot study suggests that abnormal calcitonin gene-related peptide and endothelial nitric oxide synthase activity may play a role in the development of Charcot neuroarthropathy. LEVEL OF CLINICAL EVIDENCE 4.
Collapse
Affiliation(s)
- Javier La Fontaine
- Podiatry Division, University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | | | | | | | | | | |
Collapse
|
331
|
IKK-2 is required for TNF-alpha-induced invasion and proliferation of human mesenchymal stem cells. J Mol Med (Berl) 2008; 86:1183-92. [PMID: 18600306 DOI: 10.1007/s00109-008-0378-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 05/24/2008] [Accepted: 05/27/2008] [Indexed: 01/01/2023]
Abstract
Mesenchymal stem cells (MSCs) can contribute to tissue repair by actively migrating to sites of tissue injury. However, the cellular and molecular mechanisms of MSC recruitment are largely unknown. The nuclear factor (NF)-kappaB pathway plays a pivotal role in regulating genes that influence cell migration, cell differentiation, inflammation, and proliferation. One of the major cytokines released at sites of injury is tumor necrosis factor-alpha (TNF-alpha), which is known to be a key regulator of the NF-kappaB pathway. Therefore, we hypothesized that TNF-alpha may lead to MSC invasion and proliferation by activation of the NF-kappaB pathway. TNF-receptor 1 and 2, NF-kappaB (p65), and IkappaB kinase 2 (IKK-2) are expressed in human MSCs (hMSCs). Stimulation of hMSCs with TNF-alpha caused a p65 translocation from the cytoplasm to nucleoplasm but did not change the expression profile of MSC markers. TNF-alpha strongly augmented the migration of hMSCs through the human extracellular matrix. Using lentiviral gene transfer, overexpressing a dominant-negative mutant of IKK-2 (dn-IKK-2) significantly blocked this effect. NF-kappaB target genes associated with migration (vascular cell adhesion molecule-1, CD44, and matrix metalloproteinase 9) were upregulated by TNF-alpha stimulation and blocked by dn-IKK-2. Moreover, using the bromodeoxyuridine assay, we showed that the inhibition of the NF-kappaB pathway caused a significant reduction in the basal proliferation rate. TNF-alpha stimulated the proliferation of hMSCs, whereas overexpression of dn-IKK-2 significantly blocked this effect. TNF-alpha led to the upregulated expression of the proliferation-associated gene cyclin D1. In conclusion, we demonstrated that the NF-kappaB pathway components, p65 and IKK-2, are expressed in hMSCs. Our data provide evidence that this signal transduction pathway is implicated in TNF-alpha-mediated invasion and proliferation of hMSCs. Therefore, hMSC recruitment to sites of tissue injury may, at least in part, be regulated by the NF-kappaB signal transduction pathway.
Collapse
|
332
|
Silkstone D, Hong H, Alman BA. Beta-catenin in the race to fracture repair: in it to Wnt. ACTA ACUST UNITED AC 2008; 4:413-9. [PMID: 18560386 DOI: 10.1038/ncprheum0838] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 05/08/2008] [Indexed: 01/15/2023]
Abstract
The Wnt/beta-catenin pathway regulates multiple biological events during embryonic development, including bone formation. Fracture repair recapitulates some of the processes of normal bone development, such as the formation of bone from a cartilaginous template, and many cell-signaling pathways that underlie bone development are activated during the repair process. The Wnt/beta-catenin signaling pathway is activated during fracture repair, and dysregulation of this pathway alters the normal bone-healing response. In early pluripotent mesenchymal stem cells, Wnt/beta-catenin signaling needs to be precisely regulated to facilitate the differentiation of osteoblasts; by contrast, beta-catenin is not needed for chondrocyte differentiation. Once mesenchymal stem cells are committed to the osteoblast lineage, activation of Wnt/beta-catenin signaling enhances bone formation. This activity suggests that the Wnt/beta-catenin pathway is a therapeutic target during bone repair. Indeed, treatments that activate Wnt/beta-catenin signaling, such as lithium, increase bone density and also enhance healing.
Collapse
|
333
|
Silfverswärd CJ, Sisask G, Larsson S, Ohlsson C, Frost A, Ljunggren O, Nilsson O. Bone formation in interleukin-4 and interleukin-13 depleted mice. Acta Orthop 2008; 79:410-20. [PMID: 18622847 DOI: 10.1080/17453670710015337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Cytokines play an important role in the complex process of bone formation. We have previously found an altered skeletal phenotype with reduction of cortical bone mass in mice depleted of the 2 cytokines interleukin-4 (IL-4) and interleukin-13 (IL 13). The present study was performed to investigate a potential role of IL-4 and IL-13 in fracture healing and bone induction by demineralized xenogenic bone matrix (DXBM). METHODS Callus formation in IL-4-(/)-IL-13-(/)- (IL-4/13 knockout) and wild-type (WT) male mice was compared using a standardized fracture model. The capacity of IL-4(-/-)IL-13(-/-) and WT male and female mice to form heterotopic bone was compared using intramuscular implants of DXBM. Bone formation and mechanical properties were evaluated by pQCT, ash weight, 3-point bending, radiology, and immunohistology. RESULTS In the fracture investigation substantial amounts of new bone formation by 5 weeks were found, but no differences in radiographical healing, callus volume, BMD, BMC, or mechanical properties were detected between IL-4(-/-)IL-13(-/-) and WT mice. In the DXBM investigation radiographic analysis confirmed mineralization of implants in both groups, but no difference in the amount of mineral deposition (net bone formation) between IL-4(-/-)IL-13(-/-) and WT mice was found. Immunohistology showed inhibition of autonomic nerves in the capsule of the IL-4(-/-)IL-13(-/-) group along with a lack of vascularization within the implants. INTERPRETATION Depletion of IL-4 and IL-13 does not cause any major alteration in fracture healing or heterotopic bone formation in mice. The pattern of autonomous nerve expression and expression of markers of neovascularization is, however, altered to some extent by the absence of IL-4 and IL-13.
Collapse
|
334
|
Lee DY, Cho TJ, Kim JA, Lee HR, Yoo WJ, Chung CY, Choi IH. Mobilization of endothelial progenitor cells in fracture healing and distraction osteogenesis. Bone 2008; 42:932-41. [PMID: 18326482 DOI: 10.1016/j.bone.2008.01.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2007] [Revised: 12/30/2007] [Accepted: 01/04/2008] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Fracture healing and distraction osteogenesis (DO) are unique postnatal bone formation processes, and neovascularization is critically required for successful bone regeneration. We investigated endothelial progenitor cell (EPC) mobilization during bone regeneration, and the possible contribution of EPCs to increased vascularization and new bone formation, especially in DO. METHODS Mouse tibia fracture and rat tibia DO models were used in this study. The proportion of EPCs among the peripheral and splenic mononuclear cells (MNCs) was determined by examining the endothelial lineage staining characteristics and EPC cell surface markers. Messenger RNA expression of molecules related to EPC mobilization and homing at the fracture site were analyzed by ribonuclease protection assay and reverse-transcription polymerase chain reaction. In the rat tibia DO model, we measured blood flow during DO, and determined the distribution of ex vivo-expanded and intravenously-infused EPCs. RESULTS The proportion of EPCs among the peripheral and splenic MNCs increased after fracture, peaked on post-fracture day 3, and returned to basal levels during the healing period. Messenger RNA expression of EPC mobilizing cytokines such as vascular endothelial growth factor (VEGF), stem cell factor, monocyte chemoattractant protein-1, and stromal cell-derived factor-1, were upregulated at the fracture callus. The plasma VEGF levels peaked prior to the increase in the EPC proportion. Adhesion molecules involved in EPC homing were expressed at the fracture callus. In the DO model, the temporal pattern of the increase in the EPC proportion was similar to that in the fracture healing model, but the EPC proportion increased again during the distraction and consolidation phases. The distraction gap was relatively ischemic during the distraction phase and blood flow increased profusely later in the consolidation phase. The number of EPCs homing to the bone regeneration site in the DO model correlated with the number of transplanted EPCs in a dose-dependent manner. CONCLUSIONS These findings suggest that signals from the bone regeneration site mobilize EPCs from the bone marrow into the peripheral circulation. Increased EPC mobilization and homing may contribute to neovascularization and thus to new bone formation in fracture healing and DO.
Collapse
Affiliation(s)
- Dong Yeon Lee
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, South Korea.
| | | | | | | | | | | | | |
Collapse
|
335
|
Kearns AE, Khosla S, Kostenuik PJ. Receptor activator of nuclear factor kappaB ligand and osteoprotegerin regulation of bone remodeling in health and disease. Endocr Rev 2008; 29:155-92. [PMID: 18057140 PMCID: PMC2528846 DOI: 10.1210/er.2007-0014] [Citation(s) in RCA: 567] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Accepted: 11/15/2007] [Indexed: 12/13/2022]
Abstract
Osteoclasts and osteoblasts dictate skeletal mass, structure, and strength via their respective roles in resorbing and forming bone. Bone remodeling is a spatially coordinated lifelong process whereby old bone is removed by osteoclasts and replaced by bone-forming osteoblasts. The refilling of resorption cavities is incomplete in many pathological states, which leads to a net loss of bone mass with each remodeling cycle. Postmenopausal osteoporosis and other conditions are associated with an increased rate of bone remodeling, which leads to accelerated bone loss and increased risk of fracture. Bone resorption is dependent on a cytokine known as RANKL (receptor activator of nuclear factor kappaB ligand), a TNF family member that is essential for osteoclast formation, activity, and survival in normal and pathological states of bone remodeling. The catabolic effects of RANKL are prevented by osteoprotegerin (OPG), a TNF receptor family member that binds RANKL and thereby prevents activation of its single cognate receptor called RANK. Osteoclast activity is likely to depend, at least in part, on the relative balance of RANKL and OPG. Studies in numerous animal models of bone disease show that RANKL inhibition leads to marked suppression of bone resorption and increases in cortical and cancellous bone volume, density, and strength. RANKL inhibitors also prevent focal bone loss that occurs in animal models of rheumatoid arthritis and bone metastasis. Clinical trials are exploring the effects of denosumab, a fully human anti-RANKL antibody, on bone loss in patients with osteoporosis, bone metastasis, myeloma, and rheumatoid arthritis.
Collapse
Affiliation(s)
- Ann E Kearns
- Endocrine Research Unit, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | |
Collapse
|
336
|
AI-Aql Z, Alagl A, Graves D, Gerstenfeld L, Einhorn T. Molecular Mechanisms Controlling Bone Formation during Fracture Healing and Distraction Osteogenesis. J Dent Res 2008. [DOI: 10.1177/154405910808700215 (in engl)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Fracture healing and distraction osteogenesis have important applications in orthopedic, maxillofacial, and periodontal treatment. In this review, the cellular and molecular mechanisms that regulate fracture repair are contrasted with bone regeneration that occurs during distraction osteogenesis. While both processes have many common features, unique differences are observed in the temporal appearance and expression of specific molecular factors that regulate each. The relative importance of inflammatory cytokines in normal and diabetic healing, the transforming growth factor beta superfamily of bone morphogenetic mediators, and the process of angiogenesis are discussed as they relate to bone repair. A complete summary of biological activities and functions of various bioactive factors may be found at COPE (Cytokines & Cells Online Pathfinder Encyclopedia), http://www.copewithcytokines.de/cope.cgi .
Collapse
Affiliation(s)
- Z.S. AI-Aql
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors’ Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
- Department of Orthodontics and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA; and
- Department of Preventive Dental Sciences, College of Dentistry, King Faisal University, Alkhobar, Saudi Arabia, 31952
| | - A.S. Alagl
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors’ Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
- Department of Orthodontics and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA; and
- Department of Preventive Dental Sciences, College of Dentistry, King Faisal University, Alkhobar, Saudi Arabia, 31952
| | - D.T. Graves
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors’ Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
- Department of Orthodontics and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA; and
- Department of Preventive Dental Sciences, College of Dentistry, King Faisal University, Alkhobar, Saudi Arabia, 31952
| | - L.C. Gerstenfeld
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors’ Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
- Department of Orthodontics and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA; and
- Department of Preventive Dental Sciences, College of Dentistry, King Faisal University, Alkhobar, Saudi Arabia, 31952
| | - T.A. Einhorn
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors’ Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
- Department of Orthodontics and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA; and
- Department of Preventive Dental Sciences, College of Dentistry, King Faisal University, Alkhobar, Saudi Arabia, 31952
| |
Collapse
|
337
|
AI-Aql Z, Alagl A, Graves D, Gerstenfeld L, Einhorn T. Molecular mechanisms controlling bone formation during fracture healing and distraction osteogenesis. J Dent Res 2008; 87:107-18. [PMID: 18218835 PMCID: PMC3109437 DOI: 10.1177/154405910808700215] [Citation(s) in RCA: 490] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Fracture healing and distraction osteogenesis have important applications in orthopedic, maxillofacial, and periodontal treatment. In this review, the cellular and molecular mechanisms that regulate fracture repair are contrasted with bone regeneration that occurs during distraction osteogenesis. While both processes have many common features, unique differences are observed in the temporal appearance and expression of specific molecular factors that regulate each. The relative importance of inflammatory cytokines in normal and diabetic healing, the transforming growth factor beta superfamily of bone morphogenetic mediators, and the process of angiogenesis are discussed as they relate to bone repair. A complete summary of biological activities and functions of various bioactive factors may be found at COPE (Cytokines & Cells Online Pathfinder Encyclopedia), http://www.copewithcytokines.de/cope.cgi.
Collapse
Affiliation(s)
- Z.S. AI-Aql
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors' Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
- Department of Orthodontics, Boston University School of Dental Medicine, Boston, MA 02118, USA
| | - A.S. Alagl
- Department of Preventive Dental Sciences, College of Dentistry, King Faisal University, Alkhobar, Saudi Arabia, 31952
| | - D.T. Graves
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA 02118, USA
| | - L.C. Gerstenfeld
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors' Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
| | - T.A. Einhorn
- Orthopaedic Surgical Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Doctors' Office Building, Suite 808, 720 Harrison Avenue, Boston, MA 02118, USA
| |
Collapse
|
338
|
Delos D, Yang X, Ricciardi B, Myers E, Bostrom M, Pleshko Camacho N. The effects of RANKL inhibition on fracture healing and bone strength in a mouse model of osteogenesis imperfecta. J Orthop Res 2008; 26:153-64. [PMID: 17729310 PMCID: PMC2672306 DOI: 10.1002/jor.20469] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2007] [Accepted: 05/18/2007] [Indexed: 02/04/2023]
Abstract
Currently, the standard treatment for osteogenesis imperfecta (OI) is bisphosphonate therapy. Recent studies, however, have shown delayed healing of osteotomies in a subset of OI patients treated with such agents. The current study sought to determine the effects of another therapy, RANKL inhibition, on bone healing and bone strength in the growing oim/oim mouse, a model of moderate to severe OI. Mice [73 oim/oim and 69 wild-type (WT)] were injected twice weekly with either soluble murine RANK (RANK-Fc) (1.5 mg/kg) or saline beginning at 6 weeks of age. At 8 weeks of age, the animals underwent transverse mid-diaphyseal osteotomies of the right femur. Therapy was continued until sacrifice at 2, 3, 4, or 6 weeks postfracture. At 6 weeks post-fracture, greater callus area (6.59 +/- 3.78 mm(2) vs. 2.67 +/- 2.05 mm(2), p = 0.003) and increased radiographic intensity (mineral density) (0.48 +/- 0.14 vs. 0.30 +/- 0.80, p = 0.005) were found in the RANK-Fc versus saline oim/oim group, indicating a delay in callus remodeling. Despite this delay, mechanical tests at 6 weeks postfracture revealed no significant differences in whole bone properties of stiffness and failure moment. Further, RANKL inhibition resulted in a greater failure moment and greater work to failure for the nonfractured contralateral WT bones compared to the nonfractured saline WT bones. Together, these results demonstrate that RANKL inhibition does not adversely affect the mechanical properties of healing bone in the oim/oim mice, and is associated with increased strength in intact bone in the WT mice.
Collapse
Affiliation(s)
- D. Delos
- Research Division, Hospital for Special Surgery, New York, NY
| | - X. Yang
- Research Division, Hospital for Special Surgery, New York, NY
| | - B.F. Ricciardi
- Research Division, Hospital for Special Surgery, New York, NY
| | - E.R. Myers
- Research Division, Hospital for Special Surgery, New York, NY
| | - M.P.G. Bostrom
- Research Division, Hospital for Special Surgery, New York, NY
| | | |
Collapse
|
339
|
Abstract
The Charcot syndrome is a rare complication of neuropathy in diabetes and is characterized by an acute inflammatory episode of the foot that is associated with variable degrees of dislocation, fracture, and deformity. It has no single cause but represents the final common pathway in people who are predisposed to its development by the varying overlap of several different factors. The association of the active phase of the disease with inflammation, increasing osteopenia, and increasing calcification of the arterial walls strongly suggests, however, the involvement of the receptor activator of nuclear factor-kappaB ligand (RANKL)/osteoprotegerin (OPG) cytokine pathway, which is closely involved in all three processes. The evidence for increased expression of RANKL and OPG in diabetes and neuropathy as well as its potential significance is reviewed.
Collapse
Affiliation(s)
- William Jeffcoate
- Foot Ulcer Trials Unit, Department of Diabetes and Endocrinology Nottingham University Hospitals Trust, City Hospital Campus, Nottingham, NG5 1PB, UK.
| |
Collapse
|
340
|
Yang X, Ricciardi BF, Hernandez-Soria A, Shi Y, Camacho NP, Bostrom MP. Callus mineralization and maturation are delayed during fracture healing in interleukin-6 knockout mice. Bone 2007; 41:928-36. [PMID: 17921078 PMCID: PMC2673922 DOI: 10.1016/j.bone.2007.07.022] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 06/28/2007] [Accepted: 07/03/2007] [Indexed: 11/23/2022]
Abstract
IL-6 is a pleiotropic cytokine involved in cell signaling in the musculoskeletal system, but its role in bone healing remains uncertain. The purpose of this study was to examine the role of IL-6 in fracture healing. Eight-week-old male C57BL/6 and IL-6 -/- mice were subjected to transverse, mid-diaphyseal osteotomies on the right femora. Sacrifice time points were 1, 2, 4, or 6 weeks post-fracture (N=14 per group). Callus tissue properties was analyzed by microcomputed tomography (micro-CT) and Fourier transform infrared imaging spectroscopy (FT-IRIS). Cartilage and collagen content, and osteoclast density were measured histologically. In intact unfractured bone, IL-6 -/- mice had reduced crystallinity, mineral/matrix ratio, tissue mineral density (TMD), and bone volume fraction (BVF) compared to wildtype mice. This suggests that there was an underlying deficit in baseline bone quality in IL-6 -/- mice. At 2 weeks post-fracture, the callus of IL-6 -/- mice had reduced crystallinity and mineral/matrix ratio. These changes were less evident at 4 weeks. At 2 weeks, the callus of the IL-6 -/- mice had an increased tissue mineral density (TMD), an increased cartilage and collagen content, and reduced osteoclast density compared to these parameters in wildtype mice. By 4 and 6 weeks, these parameters were no longer different between the two strains of mice. In conclusion, IL-6 -/- mice had delayed callus maturity, mineralization, and remodeling compared with the callus of the wildtype mice. These effects were transient indicating that the role of IL-6 appears to be most important in the early stages of fracture healing.
Collapse
Affiliation(s)
- Xu Yang
- The Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | | | | | - Yuexian Shi
- The Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | | | | |
Collapse
|
341
|
Kakar S, Einhorn TA, Vora S, Miara LJ, Hon G, Wigner NA, Toben D, Jacobsen KA, Al-Sebaei MO, Song M, Trackman PC, Morgan EF, Gerstenfeld LC, Barnes GL. Enhanced chondrogenesis and Wnt signaling in PTH-treated fractures. J Bone Miner Res 2007; 22:1903-12. [PMID: 17680724 DOI: 10.1359/jbmr.070724] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED Studies have shown that systemic PTH treatment enhanced the rate of bone repair in rodent models. However, the mechanisms through which PTH affects bone repair have not been elucidated. In these studies we show that PTH primarily enhanced the earliest stages of endochondral bone repair by increasing chondrocyte recruitment and rate of differentiation. In coordination with these cellular events, we observed an increased level of canonical Wnt-signaling in PTH-treated bones at multiple time-points across the time-course of fracture repair, supporting the conclusion that PTH responses are at least in part mediated through Wnt signaling. INTRODUCTION Since FDA approval of PTH [PTH(1-34); Forteo] as a treatment for osteoporosis, there has been interest in its use in other musculoskeletal conditions. Fracture repair is one area in which PTH may have a significant clinical impact. Multiple animal studies have shown that systemic PTH treatment of healing fractures increased both callus volume and return of mechanical competence in models of fracture healing. Whereas the potential for PTH has been established, the mechanism(s) by which PTH produces these effects remain elusive. MATERIALS AND METHODS Closed femoral fractures were generated in 8-wk-old male C57Bl/6 mice followed by daily systemic injections of either saline (control) or 30 microg/kg PTH(1-34) for 14 days after fracture. Bones were harvested at days 2, 3, 5, 7, 10, 14, 21, and 28 after fracture and analyzed at the tissue level by radiography and histomorphometry and at the molecular and biochemical levels level by RNase protection assay (RPA), real-time PCR, and Western blot analysis. RESULTS Quantitative muCT analysis showed that PTH treatment induced a larger callus cross-sectional area, length, and total volume compared with controls. Molecular analysis of the expression of extracellular matrix genes associated with chondrogenesis and osteogenesis showed that PTH treated fractures displayed a 3-fold greater increase in chondrogenesis relative to osteogenesis over the course of the repair process. In addition, chondrocyte hypertrophy occurred earlier in the PTH-treated callus tissues. Analysis of the expression of potential mediators of PTH actions showed that PTH treatment significantly induced the expression of Wnts 4, 5a, 5b, and 10b and increased levels of unphosphorylated, nuclear localized beta-catenin protein, a central feature of canonical Wnt signaling. CONCLUSIONS These results showed that the PTH-mediated enhancement of fracture repair is primarily associated with an amplification of chondrocyte recruitment and maturation in the early fracture callus. Associated with these cellular effects, we observed an increase in canonical Wnt signaling supporting the conclusion that PTH effects on bone repair are mediated at least in part through the activation of Wnt-signaling pathways.
Collapse
Affiliation(s)
- Sanjeev Kakar
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University Medical Center, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
342
|
Lankinen P, Mäkinen TJ, Pöyhönen TA, Virsu P, Salomäki S, Hakanen AJ, Jalkanen S, Aro HT, Roivainen A. (68)Ga-DOTAVAP-P1 PET imaging capable of demonstrating the phase of inflammation in healing bones and the progress of infection in osteomyelitic bones. Eur J Nucl Med Mol Imaging 2007; 35:352-64. [PMID: 18038133 DOI: 10.1007/s00259-007-0637-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Accepted: 10/09/2007] [Indexed: 11/28/2022]
Abstract
PURPOSE Differentiation between bacterial infection and nonbacterial inflammation remains a diagnostic challenge. Vascular adhesion protein 1 (VAP-1) is a human endothelial protein whose cell surface expression is induced under inflammatory conditions, thus making it a highly promising target molecule for studying inflammatory processes in vivo. We hypothesized that positron emission tomography (PET) with gallium-68-labeled 1,4,7,10-tetraazacyclododecane-N',N'',N''',N''''-tetraacetic acid-peptide targeted to VAP-1 ((68)Ga-DOTAVAP-P1) could be feasible for imaging the early inflammatory and infectious processes in healing bones. MATERIALS AND METHODS Thirty-four Sprague-Dawley rats with diffuse Staphylococcus aureus tibial osteomyelitis and 34 rats with healing cortical bone defects (representing the inflammation stage of healing) were PET imaged using (68)Ga-DOTAVAP-P1 as a tracer. In addition, peripheral quantitative computed tomography and conventional radiography were performed. Bone samples for quantitative bacteriology and specimens were also processed for histomorphometry of inflammatory and infectious reactions. RESULTS PET imaging showed an uptake of (68)Ga-DOTAVAP-P1 in both the osteomyelitic bones and the healing cortical bone defects during the first 36 h after surgery. Thereafter, only the osteomyelitic tibias were delineated by PET. The osteomyelitic and control animals showed a similar uptake of the (68)Ga-DOTAVAP-P1 at 24 h, whereas a significant difference was observed at 7 days (p < 0.0001). CONCLUSIONS The current study showed that PET imaging with the new (68)Ga-DOTAVAP-P1 is capable of accurately demonstrating the phase of inflammation in healing bones and the progress of bacterial infection in osteomyelitic bones. Consequently, this novel imaging agent allowed for the differentiation of bone infection due to S. aureus and normal bone healing as soon as 7 days after onset.
Collapse
Affiliation(s)
- Petteri Lankinen
- Orthopaedic Research Unit, Department of Orthopaedic Surgery and Traumatology, University of Turku, Turku, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
343
|
Abe Y, Takahata M, Ito M, Irie K, Abumi K, Minami A. Enhancement of graft bone healing by intermittent administration of human parathyroid hormone (1-34) in a rat spinal arthrodesis model. Bone 2007; 41:775-85. [PMID: 17707711 DOI: 10.1016/j.bone.2007.06.025] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 05/29/2007] [Accepted: 06/27/2007] [Indexed: 01/06/2023]
Abstract
Bone grafting is commonly used to treat skeletal disorders associated with large bone defect or unstable joint. It can take several months, however, to achieve a solid union and bony fusion sometimes delays or fails especially in osteoporosis patients. Therefore, we used a rat spinal arthrodesis model to examine whether intermittent administration of human PTH(1-34) accelerates bone graft healing. Eighty-two male Sprague-Dawley rats underwent posterolateral spinal arthrodesis surgery using autologous bone grafts. Animals were given daily subcutaneous injections of hPTH(1-34) (40 microg/kg/day PTH group) or 0.9% saline vehicle (control group) from immediately after surgery till death. Five rats each were killed 2, 4, 7, and 14 days after the surgery, and mRNA expression analysis was performed on harvested grafted bone. Seven rats each were killed 14, 28, and 42 days after the surgery, and the lumbar spine, which contained the grafted spinal segment, was subjected to fusion assessment, microstructural analysis using three-dimensional micro-computed tomography, and histologic examination. Serum bone metabolism markers were analyzed. The results indicated that PTH administration decreased the time required for graft bone healing and provided a structurally superior fusion mass in the rat spinal arthrodesis model. PTH administration increased the fusion rate on day 14 (14% in the control group and 57% in the PTH group), accelerated grafted bone resorption, and produced a larger and denser fusion mass compared to control. mRNA expression of both osteoblast- and osteoclast-related genes was upregulated by PTH treatment, and serum bone formation and resorption marker levels were higher in the PTH group than in the control group. Histologically calculated mineral apposition rate, mineralized surface and osteoclast surface were also higher in the PTH group than in the control group. These findings suggest that intermittent administration of PTH(1-34) enhanced bone turn over dominantly on bone formation at the graft site, leading to the acceleration of the spinal fusion. Based on the results of this study, intermittent injection of hPTH(1-34) might be an efficient adjuvant intervention in spinal arthrodesis surgery and all other skeletal reconstruction surgeries requiring bone grafts.
Collapse
Affiliation(s)
- Yuichiro Abe
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Kita-15 Nishi-7, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
344
|
Nikolaou V, Tsiridis E. (i) Pathways and signalling molecules. CURRENT ORTHOPAEDICS 2007; 21:249-257. [DOI: 10.1016/j.cuor.2007.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
345
|
Zhu WQ, Wang X, Wang XX, Wang ZY. Temporal and spatial expression of osteoprotegerin and receptor activator of nuclear factor -kappaB ligand during mandibular distraction in rats. J Craniomaxillofac Surg 2007; 35:103-11. [PMID: 17449257 DOI: 10.1016/j.jcms.2006.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2006] [Accepted: 12/19/2006] [Indexed: 11/16/2022] Open
Abstract
PURPOSE The expression profiles of osteoprotegerin and receptor activator of nuclear factor-kappaB ligand (RANKL) were investigated in the distraction region to reveal bone remodelling characters during mandibular distraction osteogenesis. MATERIAL AND METHODS Osteotomies were performed and external distractors were used on the mandibles of 42 adult male SD rats. After a 5-day latency period, the distractors were activated at a rate of 0.4mm/day for 6 days, followed by a 4-week consolidation period. Radiographs were taken, and specimens were harvested at the end of the latency period, when distraction was completed, and at the end of 1, 2, 3 and 4 weeks of the consolidation period. Tartrate-resistant acid phosphatase staining was used to detect activated osteoclasts. Temporospatial expression of osteoprotegerin and RANKL was investigated by using immunohistochemistry, in situ hybridization and reverse transcription polymerase chain reaction. Semi-quantitative analysis was used to characterize osteoprotegerin (OPG). RANKL and RANKL/OPG ratio. RESULTS In all time points, osteoprotegerin and RANKL were co-localized in bone marrow lining cells, osteoblasts and newly embedded osteocytes. Osteoprotegerin mRNA expression increased to a peak when distraction was completed and maintained this level until the end of week 2 of the consolidation period. RANKL mRNA expression increased steadily until the end of week 1 of the consolidation period and maintained a peak level until the end of week 3, with a slight decrease at the end of week 2. The RANKL/OPG ratio increased continuously and reached its highest level at the end of weeks 3 and 4 of the consolidation period. Tartrate-resistant acid phosphatase staining positive osteoclasts were mainly detected at weeks 2, 3 and 4 of the consolidation period in bone marrow cavities and bone surfaces. CONCLUSIONS The temporospatial expression patterns of osteoprotegerin and RANKL suggest that the osteoblast lineage cell network orchestrates bone remodelling during distraction osteogenesis and most activated bone resorption takes place during weeks 3 and 4 of the consolidation phase.
Collapse
Affiliation(s)
- Wei-Qiao Zhu
- Orthognathic Surgery Centre, Department of Oral and Maxillofacial Surgery, Peking University, School of Stomatology, South Zhongguancun Avenue 22, Beijing 100081 PR China.
| | | | | | | |
Collapse
|
346
|
Kayal RA, Tsatsas D, Bauer MA, Allen B, Al-Sebaei MO, Kakar S, Leone CW, Morgan EF, Gerstenfeld LC, Einhorn TA, Graves DT. Diminished bone formation during diabetic fracture healing is related to the premature resorption of cartilage associated with increased osteoclast activity. J Bone Miner Res 2007; 22:560-8. [PMID: 17243865 PMCID: PMC3109431 DOI: 10.1359/jbmr.070115] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
UNLABELLED Histological and molecular analysis of fracture healing in normal and diabetic animals showed significantly enhanced removal of cartilage in diabetic animals. Increased cartilage turnover was associated with elevated osteoclast numbers, a higher expression of genes that promote osteoclastogenesis, and diminished primary bone formation. INTRODUCTION Diminished bone formation, an increased incidence of nonunions, and delayed fracture healing have been observed in animal models and in patients with diabetes. Fracture healing is characterized by the formation of a stabilizing callus in which cartilage is formed and then resorbed and replaced by bone. To gain insight into how diabetes affects fracture healing, studies were carried out focusing on the impact of diabetes on the transition from cartilage to bone. MATERIALS AND METHODS A low-dose treatment protocol of streptozotocin in CD-1 mice was used to induce a type 1 diabetic condition. After mice were hyperglycemic for 3 weeks, controlled closed simple transverse fractures of the tibia were induced and fixed by intramedullary pins. Histomorphometric analysis of the tibias obtained 12, 16, and 22 days after fracture was performed across the fracture callus at 0.5 mm proximal and distal increments using computer-assisted image analysis. Another group of 16-day samples were examined by microCT. RNA was isolated from a separate set of animals, and the expression of genes that reflect the formation and removal of cartilage and bone was measured by real-time PCR. RESULTS Molecular analysis of collagen types II and X mRNA expression showed that cartilage formation was the same during the initial period of callus formation. Histomorphometric analysis of day 12 fracture calluses showed that callus size and cartilage area were also similar in normoglycemic and diabetic mice. In contrast, on day 16, callus size, cartilage tissue, and new bone area were 2.0-, 4.4-, and 1.5-fold larger, respectively, in the normoglycemic compared with the diabetic group (p < 0.05). Analysis of microCT images indicated that the bone volume in the normoglycemic animals was 38% larger than in diabetic animals. There were 78% more osteoclasts in the diabetic group compared with the normoglycemic group (p < 0.05) on day 16, consistent with the reduction in cartilage. Real-time PCR showed significantly elevated levels of mRNA expression for TNF-alpha, macrophage-colony stimulating factor, RANKL, and vascular endothelial growth factor-A in the diabetic group. Similarly, the mRNA encoding ADAMTS 4 and 5, major aggrecanases that degrade cartilage, was also elevated in diabetic animals. CONCLUSIONS These results suggest that impaired fracture healing in diabetes is characterized by increased rates of cartilage resorption. This premature loss of cartilage leads to a reduction in callus size and contributes to decreased bone formation and mechanical strength frequently reported in diabetic fracture healing.
Collapse
Affiliation(s)
- Rayyan A Kayal
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | - Dimitris Tsatsas
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | - Megan A Bauer
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | - Brian Allen
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | - Maisa O Al-Sebaei
- Department of Orthopedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sanjeev Kakar
- Department of Orthopedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Cataldo W Leone
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | - Elise F Morgan
- Department of Aerospace and Mechanical Engineering, Boston University College of Engineering, Boston, Massachusetts, USA
| | - Louis C Gerstenfeld
- Department of Orthopedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Thomas A Einhorn
- Department of Orthopedic Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Dana T Graves
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| |
Collapse
|
347
|
Abstract
Fracture healing is a complex physiological process involving a coordinated interaction of hematopoietic and immune cells within the bone marrow, in conjunction with vascular and skeletal cell precursors. Multiple factors regulate this cascade of molecular events, which affects different stages in the osteoblast and chondroblast lineage during processes such as migration, proliferation, chemotaxis, differentiation, inhibition, and extracellular protein synthesis. A clear understanding of the cellular and molecular pathways in fracture healing is not only critical for advancing fracture treatment, but it may also enhance further our knowledge of the mechanisms involved within skeletal growth and repair, as well as the mechanisms of aging. An overview of the important molecules involved in fracture healing, including osteogenic autocoids and inhibitory molecules, and their interactions and possible mechanisms of synergy during the healing process is presented in this article.
Collapse
Affiliation(s)
- Eleftherios Tsiridis
- Academic Department of Trauma and Orthopaedic Surgery, St James's University Hospital, Beckett Street, Leeds, UK
| | | | | |
Collapse
|
348
|
Cho TJ, Kim JA, Chung CY, Yoo WJ, Gerstenfeld LC, Einhorn TA, Choi IH. Expression and role of interleukin-6 in distraction osteogenesis. Calcif Tissue Int 2007; 80:192-200. [PMID: 17340223 DOI: 10.1007/s00223-006-0240-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Accepted: 11/27/2006] [Indexed: 10/23/2022]
Abstract
Distraction osteogenesis is a special form of bone healing in which well-controlled distraction stresses and consequent tensile strains within callus tissue induce very efficient new bone formation. Proinflammatory cytokines are involved during the early phase of fracture healing and callus remodeling. Temporal expression patterns of proinflammatory cytokines were assessed in Sprague-Dawley rat tibial models of distraction osteogenesis and acute lengthening, and only interleukin-6 (IL-6) was found to be specifically induced during the distraction phase. IL-6 immunoreactivity was detected not only in hemopoietic cells and osteoblasts but also in the spindle-shaped cells of the fibrous interzone, where most of the tensile strains are concentrated. In vitro study revealed that IL-6 did not affect the proliferation of C3H10T1/2 cells, mouse bone marrow stromal cells (MSCs), or MC3T3-E1 cells; but its blocking antibody reduced the proliferation of C3H10T1/2 cells and MSCs. The mRNA expression of COL1A1 and osteopontin were not changed by IL-6 or its blocking antibody, but the alkaline phosphatase activities of MC3T3-E1 cells were increased by IL-6 and decreased by its blocking antibody. These findings indicate that IL-6 is a proinflammatory cytokine that responds to tensile strain during distraction osteogenesis. IL-6 negatively affects the proliferation of primitive mesenchymal cells, whereas the differentiation of more mature osteoblastic lineage cells is enhanced by IL-6 in vitro. IL-6 appears to be one of the cytokines involved in the complex network of signal cascades evoked during distraction osteogenesis and may differentially affect immature and mature osteoblastic lineage cells.
Collapse
Affiliation(s)
- T-J Cho
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, 28 Yeongeon-dong Jongno-gu, Seoul, 110-744, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
349
|
Lories RJU, Derese I, de Bari C, Luyten FP. Evidence for uncoupling of inflammation and joint remodeling in a mouse model of spondylarthritis. ARTHRITIS AND RHEUMATISM 2007; 56:489-97. [PMID: 17265484 DOI: 10.1002/art.22372] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To study the relationship between inflammation and remodeling by inhibiting tumor necrosis factor alpha (TNFalpha) in male DBA/1 mice with spontaneous arthritis, a model of spondylarthritis (SpA). METHODS TNFalpha was inhibited using etanercept, a soluble TNF receptor. The efficacy of the dose used (25 micro g/mouse) was confirmed in methylated bovine serum albumin (mBSA)-induced monarthritis, a model of inflammation-driven joint destruction. Male DBA/1 mice with spontaneous arthritis were caged together from the age of 10 weeks onward and were treated twice weekly with etanercept. The incidence and clinical severity of disease were recorded. Mice were killed at age 25 weeks, and histomorphologic analysis was performed. The presence of TNFalpha, NF-kappaB, and Smad signaling was studied using immunohistochemistry. Entheseal endochondral bone formation was modeled using micromass cultures of periosteal cells. RESULTS Etanercept inhibited mouse TNFalpha in vitro and in vivo. Etanercept treatment of mBSA-induced arthritis had a significant effect on the severity of disease. Etanercept did not affect the incidence or severity of spontaneous arthritis. Pathologic analysis revealed no differences between etanercept-treated and phosphate buffered saline-treated mice. TNFalpha-positive cells were observed in the synovium, in vessel-associated cells, in fibrocartilage, and in new cartilage. Activation of Smad signaling was observed in earlier stages of disease than was active NF-kappaB signaling. TNFalpha inhibited chondrogenesis in the micromass model. CONCLUSION Inhibition of TNF did not affect the severity and incidence of joint ankylosis in a mouse model of SpA. Therefore, the process of entheseal ankylosis may be independent of TNF. New tissue formation in SpA could be considered an additional and specific therapeutic target.
Collapse
|
350
|
Logar DB, Komadina R, Prezelj J, Ostanek B, Trost Z, Marc J. Expression of bone resorption genes in osteoarthritis and in osteoporosis. J Bone Miner Metab 2007; 25:219-25. [PMID: 17593491 DOI: 10.1007/s00774-007-0753-0] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Accepted: 02/27/2007] [Indexed: 11/27/2022]
Abstract
Cathepsin K and MMP-9 are considered to be the most abundant proteases in osteoclasts. TRAP is a marker for osteoclasts, and there is increasing evidence of its proteolytic role in bone resorption. RANKL is a recently discovered regulator of osteoclast maturation and activity and induces expression of many genes. This study compared cathepsin K, MMP-9, TRAP, RANKL, OPG, and osteocalcin gene expression in the proximal femur of patients with osteoarthritis with that of patients with femoral neck fracture. Fifty-six patients undergoing arthroplasty because of osteoarthritis or femoral neck fracture were included in the study. Total mRNA was extracted from the bone samples obtained from the intertrochanteric region of the proximal femur. Real-time RT-PCR was used to quantify CTSK (cathepsin K), MMP-9 (matrix metalloproteinase 9), ACP5 (TRAP), TNFSF11 (RANKL), TNFRSF11B (OPG), and BGLAP (osteocalcin) mRNAs. The levels of mRNAs coding for MMP-9 and osteocalcin indicated higher expression in the osteoarthritic group (P = 0.011, P = 0.001, respectively), whereas RANKL expression and the ratio RANKL/OPG were both significantly lower in the osteoarthritic group than in the fracture group. Expression of cathepsin K, MMP-9, and TRAP relative to RANKL was significantly higher in the osteoarthritic group. Ratios of all three proteolytic enzymes relative to formation marker osteocalcin were higher in the fracture group. Gene expression of cathepsin K, MMP-9, TRAP, RANKL, OPG, and osteocalcin and the association between their mRNA levels pointed to higher bone resorption and bone formation in osteoarthritis, differences in balance between them, and differences in regulation of bone resorption in osteoarthritic and osteoporotic bone.
Collapse
Affiliation(s)
- Darja Bitenc Logar
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, SI-1000, Ljubljana, Slovenia
| | | | | | | | | | | |
Collapse
|