Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Biomaterials ; 53: 464-74, 2015.
Article in English | MEDLINE | ID: mdl-25890743

ABSTRACT

As the field of biomedical implants matures the functionality of implants is rapidly increasing. In the field of neural prostheses this is particularly apparent as researchers strive to build devices that interact with highly complex neural systems such as vision, hearing, touch and movement. A retinal implant, for example, is a highly complex device and the surgery, training and rehabilitation requirements involved in deploying such devices are extensive. Ideally, such devices will be implanted only once and will continue to function effectively for the lifetime of the patient. The first and most pivotal factor that determines device longevity is the encapsulation that separates the sensitive electronics of the device from the biological environment. This paper describes the realisation of a free standing device encapsulation made from diamond, the most impervious, long lasting and biochemically inert material known. A process of laser micro-machining and brazing is described detailing the fabrication of hermetic electrical feedthroughs and laser weldable seams using a 96.4% gold active braze alloy, another material renowned for biochemical longevity. Accelerated ageing of the braze alloy, feedthroughs and hermetic capsules yielded no evidence of corrosion and no loss of hermeticity. Samples of the gold braze implanted for 15 weeks, in vivo, caused minimal histopathological reaction and results were comparable to those obtained from medical grade silicone controls. The work described represents a first account of a free standing, fully functional hermetic diamond encapsulation for biomedical implants, enabled by gold active alloy brazing and laser micro-machining.


Subject(s)
Alloys , Biocompatible Materials , Diamond , Gold , Neural Prostheses , Zinc Oxide-Eugenol Cement , Animals , Guinea Pigs
2.
Int J Numer Method Biomed Eng ; 30(1): 1-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24039120

ABSTRACT

Denosumab, a fully human monoclonal antibody, has been approved for the treatment of postmenopausal osteoporosis. The therapeutic effect of denosumab rests on its ability to inhibit osteoclast differentiation. Here, we present a computational approach on the basis of coupling a pharmacokinetics model of denosumab with a pharmacodynamics model for quantifying the effect of denosumab on bone remodeling. The pharmacodynamics model comprises an integrated systems biology-continuum micromechanics approach, including a bone cell population model, considering the governing biochemical factors of bone remodeling (including the action of denosumab), and a multiscale micromechanics-based bone mechanics model, for implementing the mechanobiology of bone remodeling in our model. Numerical studies of postmenopausal osteoporosis show that denosumab suppresses osteoclast differentiation, thus strongly curtailing bone resorption. Simulation results also suggest that denosumab may trigger a short-term bone volume gain, which is, however, followed by constant or decreasing bone volume. This evolution is accompanied by a dramatic decrease of the bone turnover rate by more than one order of magnitude. The latter proposes dominant occurrence of secondary mineralization (which is not anymore impeded through cellular activity), leading to higher mineral concentration per bone volume. This explains the overall higher bone mineral density observed in denosumab-related clinical studies.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Computer Simulation , Models, Theoretical , Osteoporosis, Postmenopausal/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Bone Density/drug effects , Bone Remodeling/drug effects , Bone Resorption/drug therapy , Bone and Bones/drug effects , Bone and Bones/metabolism , Calibration , Denosumab , Dose-Response Relationship, Drug , Female , Humans , Postmenopause
3.
Acta Biomater ; 9(6): 7014-24, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23467040

ABSTRACT

During the past two decades, research on ceramic scaffolds for bone regeneration has progressed rapidly; however, currently available porous scaffolds remain unsuitable for load-bearing applications. The key to success is to apply microstructural design strategies to develop ceramic scaffolds with mechanical properties approaching those of bone. Here we report on the development of a unique microstructurally designed ceramic scaffold, strontium-hardystonite-gahnite (Sr-HT-gahnite), with 85% porosity, 500µm pore size, a competitive compressive strength of 4.1±0.3MPa and a compressive modulus of 170±20MPa. The in vitro biocompatibility of the scaffolds was studied using primary human bone-derived cells. The ability of Sr-HT-gahnite scaffolds to repair critical-sized bone defects was also investigated in a rabbit radius under normal load, with ß-tricalcium phosphate/hydroxyapatite scaffolds used in the control group. Studies with primary human osteoblast cultures confirmed the bioactivity of these scaffolds, and regeneration of rabbit radial critical defects demonstrated that this material induces new bone defect bridging, with clear evidence of regeneration of original radial architecture and bone marrow environment.


Subject(s)
Bone Regeneration/physiology , Bone Substitutes/chemical synthesis , Ceramics/chemistry , Guided Tissue Regeneration/instrumentation , Radius Fractures/physiopathology , Radius Fractures/surgery , Tissue Scaffolds , Animals , Equipment Design , Equipment Failure Analysis , Fracture Healing/physiology , Guided Tissue Regeneration/methods , Male , Materials Testing , Rabbits , Radius Fractures/diagnosis , Treatment Outcome
4.
Acta Biomater ; 8(11): 4162-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22842031

ABSTRACT

This is the first reported study to prepare highly porous baghdadite (Ca3ZrSi2O9) scaffolds with and without surface modification and investigate their ability to repair critical-sized bone defects in a rabbit radius under normal load. The modification was carried out to improve the mechanical properties of the baghdadite scaffolds (particularly to address their brittleness) by coating their surfaces with a thin layer (∼400 nm) of polycaprolactone (PCL)/bioactive glass nanoparticles (nBGs). The ß-tricalcium phosphate/hydroxyapatite (TCP/HA) scaffolds with and without modification were used as the control groups. All of the tested scaffolds had an open and interconnected porous structure with a porosity of ∼85% and average pore size of 500 µm. The scaffolds (six per scaffold type and size of 4 mm × 4 mm × 15 mm) were implanted (press-fit) into the rabbit radial segmental defects for 12 weeks. Micro-computed tomography and histological evaluations were used to determine bone ingrowth, bone quality, and implant integration after 12 weeks of healing. Extensive new bone formation with complete bridging of the radial defect was evident with the baghdadite scaffolds (modified/unmodified) at the periphery and in close proximity to the ceramics within the pores, in contrast to TCP/HA scaffolds (modified/unmodified), where bone tended to grow between the ulna adjacent to the implant edge. Although the modification of the baghdadite scaffolds significantly improved their mechanical properties, it did not show any significant effect on in vivo bone formation. Our findings suggest that baghdadite scaffolds with and without modification can serve as a potential material to repair critical sized bone defects.


Subject(s)
Bone and Bones/pathology , Silicates/chemistry , Tissue Scaffolds/chemistry , Wound Healing , Animals , Body Fluids , Bone Regeneration/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Calcium Phosphates/pharmacology , Disease Models, Animal , Durapatite/pharmacology , Male , Mechanical Phenomena/drug effects , Molecular Weight , Osteoclasts/metabolism , Osteoclasts/pathology , Polyesters/pharmacology , Porosity , Rabbits , Solutions , Wound Healing/drug effects , X-Ray Microtomography
6.
Int J Biochem Cell Biol ; 39(6): 1077-81, 2007.
Article in English | MEDLINE | ID: mdl-17174136

ABSTRACT

RANK ligand (RANKL), a key mediator of bone resorption in normal and pathological states, is expressed as membrane-bound or soluble forms by tissues as diverse as lymph nodes, spleen, thymus and bone-forming cells. In normal bone turnover and in bone metastasis, RANKL stimulates the formation and activity of bone-removing cells, osteoclasts, by binding to its cognate receptor, RANK, on osteoclasts and their progenitors; these processes are disrupted by binding of RANKL to osteoprotegerin (OPG), a soluble decoy receptor. Whilst no mutations in the RANKL gene have yet been identified in human disease, mutations that result in enhanced RANK signalling through inactivation of OPG or activation of RANK are associated with Juvenile Paget's disease and familial expansile osteolysis, respectively. This review focuses on the central role of RANKL in bone resorption and on the therapeutic targeting of RANKL in osteoporosis, humoral hypercalcaemia of malignancy and bone metastasis.


Subject(s)
RANK Ligand/genetics , RANK Ligand/metabolism , Amino Acid Sequence , Animals , Bone Resorption/metabolism , Bone Resorption/prevention & control , Gene Expression Profiling , Humans , Models, Biological , Molecular Sequence Data , Osteoprotegerin/metabolism , RANK Ligand/antagonists & inhibitors , Sequence Homology, Amino Acid
8.
Arthritis Rheum ; 54(6): 1772-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16736519

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is a chronic inflammatory disease that may result in debilitating joint deformities with destruction of bone and cartilage. Inflammation is still considered the pivotal inducer of both components of joint damage. Results of recent animal studies suggested a prominent contribution of osteoclastic bone resorption that could be dissociated from inflammation. RANKL and its natural decoy receptor, osteoprotegerin (OPG), play key roles in osteoclast activation. In a group of patients with early RA not treated with disease-modifying drugs, we tested the hypothesis that osteoclast activation, reflected by the serum OPG:RANKL ratio at baseline, is negatively associated with progression of bone damage, independent of inflammation. METHODS: OPG and RANKL levels, together with a parameter of inflammation (first-year time-averaged erythrocyte sedimentation rate [tESR]), were measured in 92 patients with newly diagnosed early active RA who were participants in a randomized study. The tESR and the OPG:RANKL ratio were evaluated for the ability to predict 5-year radiographic progression of joint damage. RESULTS: The first-year tESR and the OPG:RANKL ratio, as measured at baseline, independently predicted 5-year radiographic progression of joint damage (both P < or = 0.001). Progression of radiographic damage was greatest in patients with a high tESR and a low OPG:RANKL ratio and was lowest in patients with a low tESR and a high OPG:RANKL ratio. CONCLUSION: This study in patients with early untreated RA is the first to confirm the findings in animal models of arthritis, that radiographic progression of the bone component of joint destruction is dependent on both inflammation (tESR) and osteoclast activation (the OPG:RANKL ratio).


Subject(s)
Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , Carrier Proteins/blood , Glycoproteins/blood , Membrane Glycoproteins/blood , Receptors, Cytoplasmic and Nuclear/blood , Receptors, Tumor Necrosis Factor/blood , Arthritis, Rheumatoid/diagnostic imaging , Arthrography , Blood Sedimentation , Disease Progression , Female , Humans , Joints/pathology , Male , Middle Aged , Osteoclasts/physiology , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B
9.
Calcif Tissue Int ; 78(1): 35-44, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16362459

ABSTRACT

Osteoprotegerin (OPG) acts by neutralizing the receptor activator of nuclear factor-kappaB ligand (RANKL), the primary mediator of osteoclast differentiation, function, and survival. We examined whether OPG could affect the bone loss associated with chronic kidney disease (CKD) in a rodent model of CKD and secondary hyperparathyroidism (SHPT). SHPT was induced in rats by 5/6 nephrectomy (5/6 Nx) and a 1.2% P/0.6% Ca(2+) diet. Starting 1 week after 5/6 Nx, rats were treated with vehicle (veh) or OPG-Fc (3 mg/kg, intravenously) every 2 weeks for 9 weeks. At baseline, 3, 6, and 9 weeks, blood was taken and bone mineral density (BMD) and bone mineral content (BMC) were assessed by dual-energy X-ray absorptiometry. Serum parathyroid hormone (sPTH) levels reached 912 pg/ml in 5/6 Nx rats vs. 97 pg/ml in shams at 9 weeks. OPG-Fc had no effect on sPTH or Ca(2+) levels throughout the 9-week study, indicating that SHPT was a renal effect independent of bone changes. At 3 weeks, 5/6 Nx-veh rats had osteopenia compared with sham-veh rats and 5/6 Nx-OPG-Fc rats had significantly higher percent changes in whole-body BMC, leg BMD, and lumbar BMD versus 5/6 Nx-veh rats. By 6-9 weeks, elevated sPTH was associated with reversal of bone loss and osteitis fibrosa in the proximal tibial metaphysis. OPG-Fc decreased this sPTH-driven high bone turnover, resulting in augmented thickness of proximal tibial trabeculae in 5/6 Nx rats. Thus, RANKL inhibition with OPG-Fc can block the deleterious effects of continuously elevated sPTH on bone, suggesting that RANKL may be an important therapeutic target for protecting bone in patients with CKD and SHPT.


Subject(s)
Disease Models, Animal , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Hyperparathyroidism/metabolism , Kidney Failure, Chronic/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/metabolism , Absorptiometry, Photon , Animals , Carrier Proteins/antagonists & inhibitors , Glycoproteins/genetics , Humans , Hyperparathyroidism/etiology , Hyperparathyroidism/pathology , Kidney Failure, Chronic/complications , Male , Membrane Glycoproteins/antagonists & inhibitors , Osteoprotegerin , Parathyroid Hormone/blood , RANK Ligand , Rats , Rats, Sprague-Dawley , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor/genetics
10.
Bone ; 37(1): 87-95, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15869920

ABSTRACT

The combination of PTH with OPG has been proposed as a potential therapy in patients with severe osteoporosis. In the present study, we examined the bone material of aged ovariectomized (OVX) rats treated either with PTH (1-34) or OPG alone or in combination of both. The micro- and nanostructural characteristics of the mineralized bone were evaluated using quantitative backscattered electron imaging (qBEI) and small-angle X-ray scattering (SAXS). Rats (n=68) were either sham-operated or ovariectomized (OVX) at the age of 3 months, and 15 months later, OVX animals were treated either with vehicle, OPG (10 mg/kg), PTH (80 microg/kg) or a combination of both during 5.5 months. All treatments were by subcutaneous injection, 3 days per week. Secondary metaphyseal spongiosa from distal femora was assessed for mineralized bone volume (BV/TV), for the mean Ca-concentration (Camean), the width of the bone mineralization density distribution (Cawidth), as well as the average mineral particle thickness parameter (T) and the degree of alignment of the mineral particles (rho). A remarkable increase of BV/TV up to 139% (P<0.001) was observed in the PTH-treated groups independently of OPG. Camean was slightly increased (+1.7%, P<0.05) in the OPG-treated group. Cawidth was reduced (-6.4%, P<0.01, and -8.9%, P<0.001) in animals treated with OPG and PTH+OPG, respectively. In contrast, Cawidth in sham-operated rats was 16.0% (P<0.001) higher than in OVX. The T parameter was not altered in the trabecular bone within the group of treated and untreated OVX rats. However, the non-ovariectomized animals exhibited a significantly lower T value (-7.1%, P<0.01) with respect to OVX. In conclusion, qBEI and SAXS data of OVX rats suggest that PTH alone was responsible for increase of bone volume, whereas OPG positively influenced the homogeneity and density of mineralization without affecting the nanostructure of the bone material.


Subject(s)
Bone and Bones/drug effects , Calcification, Physiologic/drug effects , Glycoproteins/pharmacology , Ovariectomy , Teriparatide/pharmacology , Animals , Bone and Bones/chemistry , Bone and Bones/pathology , Calcium/analysis , Drug Therapy, Combination , Electron Probe Microanalysis , Femur/chemistry , Femur/drug effects , Femur/pathology , Glycoproteins/therapeutic use , Humans , Minerals/analysis , Minerals/chemistry , Osteoporosis/drug therapy , Osteoprotegerin , Rats , Receptors, Cytoplasmic and Nuclear/therapeutic use , Receptors, Tumor Necrosis Factor/therapeutic use , Teriparatide/therapeutic use
11.
Exp Cell Res ; 293(2): 292-301, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14729467

ABSTRACT

Receptor activator of NF-kB Ligand (RANKL) is an essential requirement for osteoclastogenesis and its activity is neutralized by binding to the soluble decoy receptor osteoprotegerin (OPG). The purpose of this work was to study the effects of RANKL and OPG during osteoclastogenesis using the murine monocytic cell line RAW 264.7 that can differentiate into osteoclasts in vitro. RAW 264.7 cells plated at 10(4) cells/cm(2) and cultured for 4 days in the presence of RANKL represent the optimal culture conditions for osteoclast differentiation, with an up-regulation of all parameters related to bone resorption: tartrate resistant acid phosphatase (TRAP), calcitonin receptor (CTR), RANK, cathepsin K, matrix metalloproteinase (MMP)-9 mRNA expressions. RANKL and OPG biological effects vary according to the differentiation state of the cells: in undifferentiated RAW 264.7 cells, TRAP expression was decreased by OPG and RANKL, RANK expression was inhibited by OPG, while MMP-9 and cathepsin K mRNA expressions were not modulated. In differentiated RAW 264.7 cells, RANKL and OPG both exert an overall inhibitory effect on the expression of all the parameters studied. In these experimental conditions, OPG-induced MMP-9 inhibition was abrogated in the presence of a blocking anti-RANKL antibody, suggesting that part of OPG effects are RANKL-dependent.


Subject(s)
Bone Remodeling/physiology , Carrier Proteins/metabolism , Cell Differentiation/physiology , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Osteoclasts/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/metabolism , Acid Phosphatase/genetics , Animals , Antibodies/pharmacology , Biomarkers , Bone Remodeling/drug effects , Carrier Proteins/antagonists & inhibitors , Cathepsin K , Cathepsins/genetics , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Line , Glycoproteins/genetics , Glycoproteins/pharmacology , Isoenzymes/genetics , Matrix Metalloproteinase 9/genetics , Membrane Glycoproteins/antagonists & inhibitors , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/drug effects , Osteoprotegerin , RANK Ligand , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Receptors, Calcitonin/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor , Reproducibility of Results , Stem Cells/drug effects , Tartrate-Resistant Acid Phosphatase
12.
Biochim Biophys Acta ; 1644(1): 1-7, 2004 Feb 02.
Article in English | MEDLINE | ID: mdl-14741739

ABSTRACT

Osteoprotegerin (OPG) is a decoy receptor for receptor activator of nuclear factor kappaB ligand (RANKL), an inducer of osteoclastogenesis via its receptor RANK. We recently demonstrated that OPG also exerts a direct effect in osteoclasts by regulating protease expression. Herein, we showed that OPG-induced pro-matrix metalloproteinase-9 activity was abolished by ras/MAPK inhibitors in purified osteoclasts. OPG induced the phosphorylation of p38 and ERK1/2 in RAW264.7 cells. Only p38 activation was totally abolished by a blocking anti-RANKL antibody or an excess of RANKL. Surface plasmon resonance experiments revealed that RANK, RANKL and OPG are able to form a tertiary complex. These results suggested a potential formation of a tertiary complex RANK-RANKL-OPG on osteoclasts. Thus, OPG is not only a soluble decoy receptor for RANKL but must be also considered as a direct effector of osteoclast functions.


Subject(s)
Glycoproteins/physiology , Osteoclasts/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/pharmacology , Cell Line , Cells, Cultured , Enzyme Activation , Glycoproteins/antagonists & inhibitors , Glycoproteins/pharmacology , Ligands , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/pharmacology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteoprotegerin , Phosphorylation , RANK Ligand , Rabbits , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Tumor Necrosis Factor , Signal Transduction/physiology , Surface Plasmon Resonance , Transfection , p38 Mitogen-Activated Protein Kinases
13.
Clin Exp Metastasis ; 21(5): 381-7, 2004.
Article in English | MEDLINE | ID: mdl-15672862

ABSTRACT

Osteoprotegerin (OPG) plays a central role in controlling bone resorption. Exogenous administration of OPG has been shown to be effective in preventing osteolysis and limiting the growth of osteolytic metastasis. The objective of this study was to investigate the effects of OPG on osteoblastic prostate cancer (CaP) metastases in an animal model. LuCaP 23.1 cells were injected intra-tibially and Fc-OPG (6.0 mg/kg) was administered subcutaneously three times a week starting either 24 hours prior to cell injection (prevention regimen) or at 4 weeks post-injection (treatment regimen). Changes in bone mineral density at the tumor site were determined by dual x-ray absorptiometry. Tumor growth was monitored by evaluating serum prostate specific antigen (PSA). Fc-OPG did not inhibit establishment of osteoblastic bone lesions of LuCaP 23.1, but it decreased growth of the tumor cells, as determined by decreases in serum PSA levels of 73.0 +/- 44.3% (P < 0.001) and 78.3 +/- 25.3% (P < 0.001) under the treatment and prevention regimens, respectively, compared to the untreated tumor-bearing animals. Administration of Fc-OPG decreased the proliferative index by 35.0% (P = 0.1838) in the treatment group, and 75.2% (P = 0.0358) in the prevention group. The results of this study suggest a potential role for OPG in the treatment of established osteoblastic CaP bone metastases.


Subject(s)
Bone Neoplasms/therapy , Glycoproteins/administration & dosage , Osteoblasts/metabolism , Prostatic Neoplasms/therapy , Receptors, Cytoplasmic and Nuclear/administration & dosage , Tibia/pathology , Animals , Bone Density , Bone Neoplasms/secondary , Injections, Subcutaneous , Lymphatic Metastasis/pathology , Male , Mice , Mice, Nude , Mice, SCID , Osteoprotegerin , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Receptors, Tumor Necrosis Factor/administration & dosage , Tibia/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Immunity ; 19(6): 849-61, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14670302

ABSTRACT

Autoimmune associated bone disease and intestinal inflammation are closely linked with deregulation and hyperactivation of autoreactive CD4 T cells. How these T cells are activated and mediate disease is not clear. Here we show that in the Interleukin 2-deficient mouse model of autoimmunity spontaneous osteopenia and colitis are caused by increased production of the ligand for receptor activator of NFkappaB (RANKL). RANKL acting via its receptor, receptor activator of NFkappaB (RANK), increases bone turnover and promotes intestinal dendritic cell (DC) survival in vivo. Modulation of RANKL-RANK interactions with exogenous recombinant osteoprotegerin (Fc-OPG) reverses skeletal abnormalities and reduces colitis by decreasing colonic DC numbers. This study identifies a common causal link between bone disease and intestinal inflammation and establishes the importance of DC in mediating colonic inflammation in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bone and Bones/drug effects , Dendritic Cells/drug effects , Glycoproteins/pharmacology , Inflammation/drug therapy , Animals , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/immunology , Bone and Bones/immunology , Carrier Proteins/metabolism , Colon/drug effects , Colon/immunology , Dendritic Cells/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inflammation/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
15.
Biochem Biophys Res Commun ; 310(3): 774-8, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-14550270

ABSTRACT

Receptor activator of NF-kappaB ligand (RANKL) is essential for osteoclast (OC) differentiation/activation and functions through its receptor RANK at the surface of the osteoclastic cells. This study investigated for the first time the direct effects of hRANKL on protease/protease inhibitor expressions and protease activities in purified rabbit osteoclast cultures, using semi-quantitative RT-PCR, gelatin zymography, and enzymatic assays. RANKL was shown to exert in vitro pro-resorptive effects by increasing osteoclast marker expressions (Tartrate resistant acid phosphatase (TRAP) and cathepsin K), MMP-9 expression, and pro-MMP-9 activity and by diminishing TIMP-1 expression, leading to an up-regulation of the MMP-9/TIMP-1 ratio.


Subject(s)
Carrier Proteins/physiology , Endopeptidases/biosynthesis , Membrane Glycoproteins/physiology , Osteoclasts/enzymology , Acid Phosphatase/biosynthesis , Animals , Cathepsin K , Cathepsins/biosynthesis , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Isoenzymes/biosynthesis , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Osteoclasts/metabolism , RANK Ligand , RNA, Messenger/metabolism , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Tartrate-Resistant Acid Phosphatase , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Up-Regulation
17.
Osteoporos Int ; 13(5): 394-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12086350

ABSTRACT

Osteoprotegerin (OPG) is a potent antiresorptive molecule that binds the final effector for osteoclastogenesis, receptor activator of NF-kappaB ligand (RANK-L). OPG production is regulated by a number of cytokines and hormones, including sex steroids, but there are few data on age and gender effects on circulating serum OPG levels, as well as possible relationships between OPG levels and bone turnover markers or bone mineral density (BMD). Thus, we measured serum OPG levels in an age-stratified, random sample of men (n = 346 age range, 23-90 years) and women (n = 304; age range 21-93 years) and related them to sex steroid levels, bone turnover markers and BMD. Serum OPG levels increased with age in both men (R = 0.39, p < 0.001) and women (R = 0.18, p < 0.01). Premenopausal women had higher OPG levels than men under age 50 years (171 +/- 6 pg/ml vs 134 +/- 6 pg/ml, respectively, p < 0.001), whereas serum OPG levels were no different in postmenopausal women compared with men = 50 years (195 +/- 7 pg/ml vs 188 +/- 7 pg/ml, respectively, p = 0.179). OPG levels correlated inversely with serum bioavailable testosterone levels in men = 50 years (R = -0.27, p < 0.001), but no associations were present with either estrogen or testosterone levels in the women. In the men, there was a trend for OPG levels to be associated positively with bone resorption markers and inversely with BMD. Collectively, the gender difference in OPG levels suggests that sex steroids may regulate OPG production in vivo, as has been found in vitro. Moreover, OPG production may also rise with increases in bone turnover, probably as a homeostatic mechanism to limit bone loss. Further studies directly testing these hypotheses should provide additional insights into the potential role of OPG in bone loss related to aging and sex steroid deficiency.


Subject(s)
Aging/blood , Glycoproteins/blood , Receptors, Cytoplasmic and Nuclear/blood , Adult , Aged , Aged, 80 and over , Bone Remodeling/physiology , Enzyme-Linked Immunosorbent Assay , Estradiol/blood , Female , Humans , Male , Middle Aged , Osteoprotegerin , Postmenopause/physiology , Premenopause/physiology , Receptors, Tumor Necrosis Factor , Sex Characteristics , Statistics, Nonparametric , Testosterone/blood
18.
Biochem Biophys Res Commun ; 293(1): 38-44, 2002 Apr 26.
Article in English | MEDLINE | ID: mdl-12054560

ABSTRACT

Cysteine proteases and matrix metalloproteinases (MMPs) are important factors in the degradation of organic matrix components of bone. Osteoprotegerin (OPG) is an osteoblast-secreted decoy receptor that inhibits osteoclast differentiation and activation. This study investigated the direct effects of human OPG on cathepsin K, MMP-9, MMP-2, and tissue inhibitors of metalloproteinases (TIMP1 and TIMP2) expressed by purified rabbit osteoclasts. The expression of two osteoclast markers, namely tartrate-resistant acid phosphatase (TRAP) and cathepsin K, was inhibited by 100 ng/mL hOPG, whereas MMP-9 expression was enhanced. Gelatinase activities were measured using a zymographic assay, and hOPG was shown to enhance both pro-MMP-9 and MMP-2 activities. Concomitantly, TIMP1 expression was greatly stimulated by hOPG, whereas TIMP2 mRNA levels were not modulated. Overall, these results show that hOPG regulates the proteases produced by purified osteoclasts differentially, producing a marked inhibitory effect on the expression of cathepsin K, the main enzyme involved in bone resorption.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Glycoproteins/physiology , Matrix Metalloproteinases/genetics , Osteoclasts/enzymology , Receptors, Cytoplasmic and Nuclear/physiology , Acid Phosphatase/genetics , Animals , Animals, Newborn , Cathepsin K , Cathepsins/genetics , Cells, Cultured , DNA Primers , Gene Expression Regulation, Enzymologic/drug effects , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Osteoprotegerin , RNA, Messenger/genetics , Rabbits , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics
19.
Blood ; 98(13): 3534-40, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739154

ABSTRACT

Multiple myeloma is a B-cell malignancy characterized by the accumulation of plasma cells in the bone marrow and the development of osteolytic bone disease. The present study demonstrates that myeloma cells express the critical osteoclastogenic factor RANKL (the ligand for receptor activator of NF-kappa B). Injection of 5T2MM myeloma cells into C57BL/KaLwRij mice resulted in the development of bone disease characterized by a significant decrease in cancellous bone volume in the tibial and femoral metaphyses, an increase in osteoclast formation, and radiologic evidence of osteolytic bone lesions. Dual-energy x-ray absorptiometry demonstrated a decrease in bone mineral density (BMD) at each of these sites. Treatment of mice with established myeloma with recombinant osteoprotegerin (OPG) protein, the soluble decoy receptor for RANKL, prevented the development of lytic bone lesions. OPG treatment was associated with preservation of cancellous bone volume and inhibition of osteoclast formation. OPG also promoted an increase in femoral, tibial, and vertebral BMD. These data suggest that the RANKL/RANK/OPG system may play a critical role in the development of osteolytic bone disease in multiple myeloma and that targeting this system may have therapeutic potential.


Subject(s)
Glycoproteins/therapeutic use , Multiple Myeloma/complications , Osteolysis/prevention & control , Receptors, Cytoplasmic and Nuclear/therapeutic use , Animals , Bone Density , Bone and Bones/pathology , Carrier Proteins/analysis , Carrier Proteins/genetics , Flow Cytometry , Gene Expression , Glycoproteins/administration & dosage , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Multiple Myeloma/chemistry , Multiple Myeloma/pathology , Neoplasm Transplantation , Osteolysis/etiology , Osteolysis/pathology , Osteoprotegerin , RANK Ligand , RNA, Messenger/analysis , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear/administration & dosage , Receptors, Tumor Necrosis Factor , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction
20.
Clin Cancer Res ; 7(10): 2977-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595685

ABSTRACT

PURPOSE: Osteoprotegerin (OPG) is a soluble osteoclastogenesis inhibitor that regulates bone turnover. We reported recently that OPG protein expression is significantly increased in prostate cancer (CaP) cells present in bone metastases. The aim of this study was to determine serum OPG levels in patients at different stages of CaP and correlate the results with disease status. EXPERIMENTAL DESIGN: OPG levels were examined in patients with benign prostatic hyperplasia, clinically localized CaP, early recurrence of CaP, and advanced CaP and evidence of bone metastases. Serum OPG levels were measured by sandwich ELISA assays. The serum Crosslaps (sCTX) assay was used to quantify bone resorption in the advanced CaP group. RESULTS: Serum OPG levels were increased significantly in the advanced CaP group versus all other groups. There was no significant correlation between serum OPG levels and PSA levels either in the advanced CaP group or within any of three treatment subclasses of this group: no Tx, those not treated; Tx, those treated; and R, those treated with resorption blockers. Levels of OPG were negatively correlated with sCTX levels only in the advanced CaP Tx group. sCTX levels correlated with prostate-specific antigen levels in the advanced CaP Tx and R groups but not in the no-Tx group. CONCLUSIONS: Our data show that serum OPG levels are increased with advanced CaP. We hypothesize that OPG levels are related to CaP progression and suggest that further studies of the biological effects of OPG on CaP metastases are warranted.


Subject(s)
Glycoproteins/blood , Prostatic Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/blood , Adult , Aged , Aged, 80 and over , Bone Neoplasms/blood , Bone Neoplasms/secondary , Collagen/blood , Enzyme-Linked Immunosorbent Assay , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/pathology , Osteoprotegerin , Peptide Fragments/blood , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/blood , Receptors, Tumor Necrosis Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...