Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Dent Res ; 97(9): 1031-1038, 2018 08.
Article in English | MEDLINE | ID: mdl-29617179

ABSTRACT

Alveolar bone is a mechanosensitive tissue that provides structural support for teeth. Alveolar bone loss is common with aging, menopause, tooth loss, and periodontitis and can lead to additional tooth loss, reduced denture fixation, and challenges in placing dental implants. The current studies suggest that sclerostin and DKK1, which are established osteocyte-derived inhibitors of bone formation, contribute to alveolar bone loss associated with estrogen ablation and edentulism in rats. Estrogen-deficient ovariectomized rats showed significant mandibular bone loss that was reversed by systemic administration of sclerostin antibody (SAB) alone and in combination with DKK1 antibody (DAB). Osteocytes in the dentate and edentulous rat maxilla expressed Sost (sclerostin) and Dkk1 (DKK1) mRNA, and molar extraction appeared to acutely increase DKK1 expression. In a chronic rat maxillary molar extraction model, systemic SAB administration augmented the volume and height of atrophic alveolar ridges, effects that were enhanced by coadministering DAB. SAB and SAB+DAB also fully reversed bone loss that developed in the opposing mandible as a result of hypo-occlusion. In both treatment studies, alveolar bone augmentation with SAB or SAB+DAB was accompanied by increased bone mass in the postcranial skeleton. Jaw bone biomechanics showed that intact sclerostin-deficient mice exhibited stronger and denser mandibles as compared with wild-type controls. These studies show that sclerostin inhibition, with and without DKK1 coinhibition, augmented alveolar bone volume and architecture in rats with alveolar bone loss. These noninvasive approaches may have utility for the conservative augmentation of alveolar bone.


Subject(s)
Alveolar Bone Loss/drug therapy , Alveolar Ridge Augmentation/methods , Bone Morphogenetic Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Absorptiometry, Photon , Alveolar Bone Loss/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Female , Genetic Markers , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice, Knockout , Ovariectomy , Phenotype , Rats , Rats, Sprague-Dawley , Tooth Extraction , X-Ray Microtomography
2.
Osteoporos Int ; 29(3): 685-697, 2018 03.
Article in English | MEDLINE | ID: mdl-29260289

ABSTRACT

Abaloparatide, a novel PTH1 receptor agonist, increased bone formation in osteopenic ovariectomized cynomolgus monkeys while increasing cortical and trabecular bone mass. Abaloparatide increased bone strength and maintained or enhanced bone mass-strength relationships, indicating preserved or improved bone quality. INTRODUCTION: Abaloparatide is a selective PTH1R activator that is approved for the treatment of postmenopausal osteoporosis. The effects of 16 months of abaloparatide administration on bone formation, resorption, density, and strength were assessed in adult ovariectomized (OVX) cynomolgus monkeys (cynos). METHODS: Sixty-five 9-18-year-old female cynos underwent OVX surgery, and 15 similar cynos underwent sham surgery. After a 9-month period without treatments, OVX cynos were allocated to four groups that received 16 months of daily s.c. injections with either vehicle (n = 17) or abaloparatide (0.2, 1, or 5 µg/kg/day; n = 16/dose level), while Sham controls received s.c. vehicle (n = 15). Bone densitometry (DXA, pQCT, micro-CT), qualitative bone histology, serum calcium, bone turnover markers, bone histomorphometry, and bone strength were among the key measures assessed. RESULTS: At the end of the 9-month post-surgical bone depletion period, just prior to the treatment phase, the OVX groups exhibited increased bone turnover markers and decreased bone mass compared with sham controls. Abaloparatide administration to OVX cynos led to increased bone formation parameters, including serum P1NP and endocortical bone formation rate. Abaloparatide administration did not influence serum calcium levels, bone resorption markers, cortical porosity, or eroded surfaces. Abaloparatide increased bone mass at the whole body, lumbar spine, tibial diaphysis, femoral neck, and femoral trochanter. Abaloparatide administration was associated with greater lumbar vertebral strength, and had no adverse effects on bone mass-strength relationships for the vertebrae, femoral neck, femoral diaphysis, or humeral cortical beams. CONCLUSIONS: Abaloparatide administration was associated with increases in bone formation, bone mass and bone strength, and with maintenance of bone quality in OVX cynos, without increases in serum calcium or bone resorption parameters.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/prevention & control , Osteogenesis/drug effects , Parathyroid Hormone-Related Protein/therapeutic use , Absorptiometry, Photon/methods , Animals , Biomarkers/blood , Bone Density/drug effects , Bone Density/physiology , Bone Density Conservation Agents/pharmacology , Bone Resorption/physiopathology , Female , Lumbar Vertebrae/physiopathology , Macaca fascicularis , Osteogenesis/physiology , Ovariectomy , Parathyroid Hormone-Related Protein/pharmacology , Peptide Fragments/blood , Procollagen/blood , Receptor, Parathyroid Hormone, Type 1/agonists , X-Ray Microtomography/methods
3.
Horm Metab Res ; 43(13): 944-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22161252

ABSTRACT

Inhibition of the receptor activator of NF-κB ligand (RANKL) is a novel therapeutic option in the treatment of osteoporosis and related diseases. The aim of this study was to evaluate bone metabolism and structure in pigs after RANKL inhibition. 12 growing pigs were assigned to 2 groups with 6 animals each. The OPG group received recombinant human OPG-Fc (5 mg/kg IV) at day 0, the control group was given 0.9% NaCl solution. Serum levels of OPG-Fc, calcium (Ca), phosphorus (P), and bone turnover markers were evaluated every 5 days, and pigs were euthanized on day 20. Serum OPG-Fc concentration peaked at day 5 and coincided with significantly decreased Ca, P, and bone turnover markers. By day 15, measureable OPG-Fc serum levels could only be detected in 2/6 animals. With OPG-Fc clearance starting at day 10, serum Ca and P concentrations were not different between the 2 groups. TRACP5b, P1CP, and BAP levels significantly decreased by 40-70% relative to vehicle controls in the OPG-Fc group between days 5 and 10, indicating that pharmacologic concentration of OPG-Fc led to systemic concomitant inhibition of bone formation and resorption in young growing pigs. Dual X-ray absorptiometry data derived from the proximal femur did not differ between the 2 groups. µCT analysis of selected bone sites demonstrated an OPG-Fc-induced improvement of specific bone architectural indices and bone mineralization.


Subject(s)
Bone Resorption , Bone and Bones/metabolism , Calcium/blood , Immunoglobulin Fc Fragments/pharmacology , Osteoprotegerin/pharmacology , Absorptiometry, Photon , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Bone and Bones/diagnostic imaging , Female , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Male , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Phosphorus/blood , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Swine
4.
Calcif Tissue Int ; 82(5): 361-72, 2008 May.
Article in English | MEDLINE | ID: mdl-18465074

ABSTRACT

Receptor activator for nuclear factor-kappa B ligand (RANKL) is an essential mediator of osteoclastogenesis. We hypothesized that administration of soluble RANKL to mice would result in high turnover and deleterious effects on both cortical and trabecular bone. For 10 days, 10-week-old C57BL/6J female mice (n = 12/group) were given twice-daily subcutaneous injections of human recombinant RANKL (0.4 or 2 mg/kg/day) or inert vehicle (VEH). Bone turnover was greatly accelerated by RANKL, as evidenced by the 49-84% greater levels of serum TRAP-5b (bone resorption marker) and 300-400% greater levels of serum alkaline phosphatase (bone formation marker). RANKL resulted in significantly greater endocortical bone erosion surface (79-83%) and periosteal bone formation rate (64-87%) vs. VEH. Microcomputed tomographic (microCT) analysis of the proximal tibia indicated a reduction in trabecular volume fraction (-84%) for both doses of RANKL. Cortical bone geometry and strength were also negatively influenced by RANKL. MicroCT analysis of the femoral diaphysis indicated significantly lower cortical bone volume (-10% to -13%) and greater cortical porosity (8-9%) relative to VEH. Biomechanical testing of the femur diaphysis revealed significantly lower maximum bending load (-19% to -25%) vs. VEH. Bone strength remained correlated with bone mass, independent of RANKL stimulation of bone turnover. These findings are consistent with the hypothesis that soluble RANKL could be an important etiologic factor in pathologic bone loss. RANKL also has potential utility as a model for studying the consequences of high bone turnover on bone quality and strength in animals.


Subject(s)
Bone Density/drug effects , Bone Remodeling/drug effects , Femur/drug effects , RANK Ligand/pharmacology , Tibia/drug effects , Acid Phosphatase/blood , Animals , Biomarkers/blood , Bone Density/physiology , Bone Remodeling/physiology , Compressive Strength/drug effects , Dose-Response Relationship, Drug , Female , Femur/diagnostic imaging , Femur/metabolism , Humans , Injections, Subcutaneous , Isoenzymes/blood , Mice , Mice, Inbred C57BL , Recombinant Proteins , Tartrate-Resistant Acid Phosphatase , Tibia/diagnostic imaging , Tibia/metabolism , Tomography, X-Ray Computed
5.
Osteoporos Int ; 19(5): 625-35, 2008 May.
Article in English | MEDLINE | ID: mdl-18038244

ABSTRACT

UNLABELLED: RANKL was administered continuously to rats for 28 days to investigate its potential as a disease model for the skeletal system. Bone turnover rates, bone material, structural and mechanical properties were evaluated. RANKL infusion caused overall skeletal complications comparable to those in high bone-turnover conditions, such as postmenopausal osteoporosis. INTRODUCTION: RANKL is an essential mediator for osteoclast development. No study has examined in detail the direct skeletal consequences of excess RANKL on bone turnover, mineralization, architecture, and vascular calcification. We, therefore, administrated soluble RANKL continuously into mature rats and created a bone-loss model. METHODS: Six-month-old Sprague-Dawley (SD) rats were assigned to three groups (n = 12) receiving continuous administration of saline (VEH) or human RANKL (35 microg/kg/day, LOW or 175 microg/kg/day, HI) for 28 days. Blood was collected routinely during the study. At sacrifice, hind limbs and aorta were removed and samples were analyzed. RESULTS: High dose RANKL markedly stimulated serum osteocalcin and TRAP-5b levels and reduced femur cortical bone volume (-7.6%) and trabecular volume fraction (BV/TV) at the proximal tibia (-64% vs. VEH). Bone quality was significantly degraded in HI, as evidenced by decreased femoral percent mineralization, trabecular connectivity, and increased endocortical bone resorption perimeters. Both cortical and trabecular bone mechanical properties were reduced by high dose RANKL. No differences were observed in the mineral content of the abdominal aorta. CONCLUSIONS: Continuous RANKL infusion caused general detrimental effects on rat skeleton. These changes are comparable to those commonly observed in high-turnover bone diseases such as postmenopausal osteoporosis.


Subject(s)
Bone Density/drug effects , Bone Remodeling/drug effects , Disease Models, Animal , Osteoporosis/chemically induced , RANK Ligand/pharmacology , Animals , Biomarkers/blood , Male , Osteoporosis/pathology , Rats , Rats, Sprague-Dawley
6.
Osteoporos Int ; 18(8): 1073-82, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17384898

ABSTRACT

UNLABELLED: Weekly treatment of gonad-intact cynomolgus monkeys (for up to 6 months) with the RANKL inhibitor OPG-Fc reduced bone turnover markers and increased volumetric cortical and trabecular BMD and BMC at radial and tibial metaphyses. OPG-Fc was well tolerated in this study without evidence of change in measured toxicologic parameters vs. control. INTRODUCTION: RANKL is the primary mediator of osteoclast formation, function, and survival. The catabolic effects of RANKL are inhibited by OPG, a soluble decoy receptor for RANKL. We investigated the safety and pharmacology of OPG-Fc in gonad-intact cynomolgus monkeys. METHODS: Males and females were treated weekly with vehicle (n = 5/sex) or OPG-Fc (15 mg/kg) by s.c. (n = 5/sex) or i.v. (n = 3/sex) injection for 6 months. RESULTS: Routine toxicologic investigations, hematologic parameters, body and organ weights, and ophthalmologic and electrocardiographic findings were not affected by OPG-Fc treatment. Because s.c. and i.v. dosing of OPG-Fc caused similar effects, these groups were combined for analyses. The following endpoints were significantly different in males and/or females treated with OPG-Fc relative to sex-matched vehicle controls after 6 months (p < 0.05). Biochemical markers of bone turnover (urine N-telopeptide and serum osteocalcin) were significantly decreased with OPG-Fc treatment. Cortical and trabecular volumetric BMD and BMC, cortical thickness, and cross-sectional moment of inertia were significantly increased by OPG-Fc treatment at the proximal tibia and distal radius metaphyses. Increases in cortical thickness were associated with significantly greater periosteal circumference. CONCLUSIONS: OPG-Fc increased cortical and trabecular BMD and BMC in young gonad-intact cynomolgus monkeys.


Subject(s)
Bone and Bones/drug effects , Osteoprotegerin/administration & dosage , RANK Ligand/antagonists & inhibitors , Receptors, IgG/administration & dosage , Animals , Bone Density/drug effects , Bone Remodeling/drug effects , Bone and Bones/pathology , Drug Administration Schedule , Female , Gonads/physiology , Macaca fascicularis , Male , Osteoprotegerin/adverse effects , Parathyroid Hormone/blood , Radius/drug effects , Radius/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Sex Factors , Tibia/drug effects , Tibia/pathology
7.
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
8.
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
9.
Gut ; 54(1): 78-86, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15591508

ABSTRACT

BACKGROUND AND AIMS: Transfer of CD4+CD45RBHi T cells into semi syngeneic immunodeficient mice represents a model of inflammatory bowel disease (IBD). As patients with IBD often suffer from osteopenia, we studied if this T cell transfer in mice results in osteopenia in addition to colitis, and if treatment with osteoprotegerin (OPG) has effects on the bone mineral density of T cell transferred mice. We also investigated whether osteopenia was due to malabsorption as a result of a dysregulated digestive tract or as a consequence of the inflammatory process. METHODS: CD4+CD45RBHi or CD4+CD45RBLo T cells (4 x 10(5)) were sorted from CB6F1 and transferred into C.B.17 scid/scid mice. Recipient mice were treated with human IgG1 Fc (control) or Fc-OPG three times per week in a prophylactic regimen as well as a therapeutic regimen (after 10% body weight loss) and were evaluated for osteopenia and colitis. RESULTS: Mice that received CD4+CD45RBHi T cells developed osteopenia (as indicated by decreased bone density accompanied by decreased osteoblasts and increased osteoclasts) and colitis (as indicated by histological changes in the large intestine). Mice that received CD4+CD45RBLo T cells developed neither osteopenia nor colitis. All animals consumed, on average, the same amount of food and water over the course of the study. Prophylactic treatment with Fc-OPG increased bone density in mice that received either CD4+CD45RBHi or CD4+CD45RBLo T cells but had no effects on the gastrointestinal tract. Fc-OPG treatment of osteopenic mice with established IBD caused the normalisation of bone density. Osteopenia in CD4+CD45RBHi T cell recipients was accompanied by hypoparathyroidism that was partially normalised by treatment with Fc-OPG. CD4+CD45RBHi T cell recipients also had a bone marrow inflammatory cell infiltrate expressing tumour necrosis factor alpha which was unaffected by treatment with Fc-OPG. CONCLUSIONS: CD4+CD45RBHi T cell transfer results in osteopenia in addition to colitis. Evidence suggests that this osteopenia was induced by inflammatory cell infiltration and not by malabsorption of calcium. Recombinant human osteoprotegerin effectively treated the osteopenia. OPG may be a useful therapeutic option for treating osteopenia in patients with IBD.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Glycoproteins/therapeutic use , Inflammatory Bowel Diseases/complications , Lymphocyte Transfusion/adverse effects , Receptors, Cytoplasmic and Nuclear/therapeutic use , Animals , Bone Density/drug effects , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/etiology , CD4-Positive T-Lymphocytes/transplantation , Female , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestine, Large/pathology , Mice , Mice, SCID , Osteoblasts/pathology , Osteoclasts/pathology , Osteoprotegerin , Parathyroid Hormone/blood , Receptors, Tumor Necrosis Factor , Recombinant Proteins/therapeutic use , Serum Amyloid A Protein/metabolism , T-Lymphocyte Subsets/transplantation , Weight Loss
11.
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
12.
J Appl Physiol (1985) ; 95(6): 2462-70, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12882990

ABSTRACT

To investigate the effects of microgravity on murine skeletal muscle fiber size, muscle contractile protein, and enzymatic activity, female C57BL/6J mice, aged 64 days, were divided into animal enclosure module (AEM) ground control and spaceflight (SF) treatment groups. SF animals were flown on the space shuttle Endeavour (STS-108/UF-1) and subjected to approximately 11 days and 19 h of microgravity. Immunohistochemical analysis of muscle fiber cross-sectional area revealed that, in each of the muscles analyzed, mean muscle fiber cross-sectional area was significantly reduced (P < 0.0001) for all fiber types for SF vs. AEM control. In the soleus, immunohistochemical analysis of myosin heavy chain (MHC) isoform expression revealed a significant increase in the percentage of muscle fibers expressing MHC IIx and MHC IIb (P < 0.05). For the gastrocnemius and plantaris, no significant changes in MHC isoform expression were observed. For the muscles analyzed, no alterations in MHC I or MHC IIa protein expression were observed. Enzymatic analysis of the gastrocnemius revealed a significant decrease in citrate synthase activity in SF vs. AEM control.


Subject(s)
Adaptation, Physiological/physiology , Muscle, Skeletal/physiology , Weightlessness/adverse effects , Animals , Body Weight/physiology , Citrate (si)-Synthase/metabolism , Female , Heart/anatomy & histology , Immunohistochemistry , Isomerism , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/metabolism , Organ Size/physiology , Oxidation-Reduction , Space Flight
13.
Matrix Biol ; 22(3): 241-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12853034

ABSTRACT

Skeletal homeostasis is partly regulated by the mechanical environment and specific signals generated by a cell's adhesion to the matrix. Previous studies demonstrated that osteopontin (OPN) expression is stimulated in response to both cellular adhesion and mechanical stimulation. The present studies examine if specific integrin ligands mediate osteoblast selective adhesion and whether opn mRNA expression is induced in response to these same ligands. Embryonic chicken calvaria osteoblastic cells were plated on bacteriological dishes coated with fibronectin (FN), collagen type I (Col1), denatured collagen/gelatin (G), OPN, vitronectin (VN), laminin (LN) or albumin (BSA). Osteoblastic cells were shown to selectively adhere to FN, Col1, G and LN, yet not to VN, OPN or BSA. Opn mRNA expression was induced by adhesion to Col1, FN, LN and G, but neither OPN nor VN induced this expression. Examination of the activation of the protein kinases A and C second signaling systems showed that only adhesion to FN induced protein kinase A and protein kinase C (PKC) activity while adherence to Col1 induced PKC. Evaluation of the intracellular distribution of focal adhesion kinase (FAK) and p-tyrosine within cells after adherence to FN, VN or BSA demonstrated that adherence to FN stimulated FAK translocation from the nucleus to the cytoplasm and high levels of p-tyrosine localization at the cell surface. However, cell adherence to VN or BSA did not show these morphological changes. These data illustrate that osteoblast selective adhesion is mediated by specific integrin ligands, and induction of intracellular second signal kinase activity is related to the nature of the ligand.


Subject(s)
Integrins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Sialoglycoproteins/genetics , Animals , Cell Adhesion , Chick Embryo , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Enzyme Induction , Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Fibronectins/pharmacology , Gene Expression Regulation , Ligands , Osteoblasts/drug effects , Osteopontin , Phosphotyrosine/metabolism , Protein Kinase C/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
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
15.
Endocrinology ; 142(10): 4295-304, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564687

ABSTRACT

PTH is a potent bone anabolic factor, and its combination with antiresorptive agents has been proposed as a therapy for osteoporosis. We tested the effects of PTH, alone and in combination with the novel antiresorptive agent OPG, in a rat model of severe osteopenia. Sprague Dawley rats were sham-operated or ovariectomized at 3 months of age. Rats were untreated for 15 months, at which time ovariectomy had caused significant decreases in bone mineral density in the lumbar vertebrae and femur. Rats were then treated for 5.5 months with vehicle (PBS), human PTH-(1-34) (80 microg/kg), rat OPG (10 mg/kg), or OPG plus PTH (all three times per wk, sc). Treatment of ovariectomized rats with OPG or PTH alone increased bone mineral density in the lumbar vertebrae and femur, whereas PTH plus OPG caused significantly greater and more rapid increases than either therapy alone (P < 0.05). OPG significantly reduced osteoclast surface in the lumbar vertebrae and femur (P < 0.05 vs. sham or ovariectomized), but had no effect on osteoblast surface at either site. Ovariectomy significantly decreased the mechanical strength of the lumbar vertebrae and femur. In the lumbar vertebrae, OPG plus PTH was significantly more effective than PTH alone at reversing ovariectomy-induced deficits in stiffness and elastic modulus. These data suggest that OPG plus PTH represent a potentially useful therapeutic option for patients with severe osteoporosis.


Subject(s)
Bone Diseases, Metabolic/drug therapy , Glycoproteins/pharmacology , Peptide Fragments/pharmacology , Teriparatide/pharmacology , Animals , Bone Density/drug effects , Bone Diseases, Metabolic/physiopathology , Drug Interactions , Drug Therapy, Combination , Female , Glycoproteins/therapeutic use , Osteoprotegerin , Ovariectomy , Peptide Fragments/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/therapeutic use , Receptors, Tumor Necrosis Factor , Teriparatide/analogs & derivatives , Teriparatide/therapeutic use
16.
Cancer Res ; 61(11): 4432-6, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389072

ABSTRACT

Certain malignancies, including breast cancer, frequently metastasize to bone, where the tumor cells induce osteoclasts to locally destroy bone. Osteoprotegerin (OPG), a member of the tumor necrosis factor receptor family, is a negative regulator of osteoclast differentiation, activation, and survival. We tested the ability of recombinant OPG to inhibit tumor-induced osteoclastogenesis, osteolysis, and skeletal tumor burden in two animal models. In a syngeneic model, mouse colon adenocarcinoma (Colon-26) cells were injected into the left ventricle of mice. Treatment with OPG dose-dependently decreased the number and area of radiographically evident lytic bone lesions, which, at the highest dose, were undetectable. Histologically, OPG also decreased skeletal tumor burden and tumor-associated osteoclasts. In a nude mouse model, OPG treatment completely prevented radiographic osteolytic lesions caused by human MDA-MB-231 breast cancer cells. Histologically, OPG decreased skeletal tumor burden by 75% and completely eradicated MDA tumor-associated osteoclasts. In both models, OPG had no effect on metastatic tumor burden in a panel of soft tissue organs. These data indicate that OPG may be an effective therapy for preventing osteolysis and decreasing skeletal tumor burden in patients with bone metastasis.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Glycoproteins/pharmacology , Osteolysis/drug therapy , Adenocarcinoma/pathology , Animals , Bone Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Transformed , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Nude , Osteoprotegerin , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor , Xenograft Model Antitumor Assays
17.
Curr Pharm Des ; 7(8): 613-35, 2001 May.
Article in English | MEDLINE | ID: mdl-11375772

ABSTRACT

OPG is a new member of the tumor necrosis factor (TNF) receptor family which plays a key role in the physiological regulation of osteoclastic bone resorption. The protein, which is produced by osteoblasts and marrow stromal cells, lacks a transmembrane domain and acts as a secreted decoy receptor which has no direct signaling capacity. OPG acts by binding to its natural ligand OPGL, which is also known as RANKL (receptor activator of NF-kappaB ligand). This binding prevents OPGL from activating its cognate receptor RANK, which is the osteoclast receptor vital for osteoclast differentiation, activation and survival. Overexpression of OPG in transgenic mice leads to profound osteopetrosis secondary to a near total lack of osteoclasts. Conversely, ablation of the OPG gene causes severe osteoporosis in mice. Ablation of OPGL or RANK also produces profound osteopetrosis, indicating the important physiological role of these proteins in regulating bone resorption. The secretion of OPG and OPGL from osteoblasts and stromal cells is regulated by numerous hormones and cytokines, often in a reciprocal manner. The relative levels of OPG and OPGL production are thought to ultimately dictate the extent of bone resorption. Excess OPGL increases bone resorption, whereas excess OPG inhibits resorption. Recombinant OPG blocks the effects of virtually all factors which stimulate osteoclasts, in vitro and in vivo. OPG also inhibits bone resorption in a variety of animal disease models, including ovariectomy-induced osteoporosis, humoral hypercalcemia of malignancy, and experimental bone metastasis. OPG might represent an effective therapeutic option for diseases associated with excessive osteoclast activity.


Subject(s)
Bone Resorption/prevention & control , Glycoproteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Bone Remodeling , Carrier Proteins/physiology , Gene Expression Regulation , Glycoproteins/chemistry , Glycoproteins/genetics , Humans , Membrane Glycoproteins/physiology , Mice , Mice, Transgenic , Osteoclasts/physiology , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor
18.
Mol Ther ; 3(2): 197-205, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11237676

ABSTRACT

Osteoprotegerin (OPG) regulates bone resorption by inhibiting osteoclast formation, function, and survival. The current studies employed a mouse ovariectomy (OVX) model of estrogen deficiency to investigate gene therapy with OPG as a means of preventing osteoporosis. Young adult females injected with a recombinant adenoviral (Ad) vector carrying cDNA of either full-length OPG or a fusion protein combining the hOPG ligand-binding domain with the human immunoglobulin constant domain (Ad-hOPG-Fc) developed serum OPG concentrations exceeding the threshold needed for efficacy. However, elevated circulating OPG levels were sustained for up to 18 months only in mice given Ad-hOPG-Fc. Administration of Ad-hOPG-Fc titers between 10(7) and 10(9) pfu yielded dose-dependent increases in serum OPG. Mice subjected to OVX or sham surgery followed by immediate treatment with Ad-hOPG-Fc had significantly more bone volume with reduced osteoclast numbers in axial and appendicular bones after 4 weeks. In contrast, animals given OVX and either a control vector or vehicle had significantly less bone than did comparably treated sham-operated mice. This study demonstrates that a single adenoviral gene transfer can produce persistent high-level OPG expression and shows that gene therapy to provide sustained delivery of OPG may prove useful in treating osteoporosis.


Subject(s)
Adenoviridae/genetics , Glycoproteins/genetics , Osteoporosis/therapy , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Biological Assay , Blotting, Southern , Blotting, Western , Bone Density/drug effects , Bone Resorption , DNA, Complementary/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Humans , Ligands , Mice , Mice, Inbred C57BL , Osteoprotegerin , Ovariectomy , Ovary/physiology , Pelvis/diagnostic imaging , Radiography , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/metabolism , Time Factors
19.
J Mater Sci Mater Med ; 12(7): 583-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-15348250

ABSTRACT

Osteoprotegerin (OPG) is a naturally secreted protein that decreases bone resorption by inhibiting osteoclast differentiation and activation while promoting osteoclast apoptosis [8]. In this study, the effects of osteoprotegerin injections on long bone mechanical and material properties were investigated in young male Sprague-Dawley rats. OPG increased fracture strength at the femur mid-diaphysis in three-point bending by 30%, without affecting the elastic or maximum strength. At the femoral neck, OPG significantly increased the elastic (45%), maximum (15%), and fracture (35%) strengths. There was not a difference in microhardness at the femur mid-diaphysis in comparing the placebo and OPG groups. There were, however, significant increases in whole bone dry mass (25%), mineral mass (30%), organic mass (17%), and percent mineralization (4%); percent mineralization at the mid-diaphysis (3%); and percent mineralization at the distal epiphysis (6%) due to the OPG treatment. While OPG decreased endocortical bone formation (52%), total bone area, endocortical bone area, and periosteal bone formation were maintained with OPG treatment. A 30% increase in the X-ray opacity of the bone at the proximal metaphysis of the right tibiae was observed. Overall, OPG increased mineralization and strength indices in the rat femur. Its effects on strength were more pronounced in the femoral neck than at the mid-diaphysis.

20.
J Exp Med ; 192(4): 463-74, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10952716

ABSTRACT

High systemic levels of osteoprotegerin (OPG) in OPG transgenic mice cause osteopetrosis with normal tooth eruption and bone elongation and inhibit the development and activity of endosteal, but not periosteal, osteoclasts. We demonstrate that both intravenous injection of recombinant OPG protein and transgenic overexpression of OPG in OPG(-/-) mice effectively rescue the osteoporotic bone phenotype observed in OPG-deficient mice. However, intravenous injection of recombinant OPG over a 4-wk period could not reverse the arterial calcification observed in OPG(-/-) mice. In contrast, transgenic OPG delivered from mid-gestation through adulthood does prevent the formation of arterial calcification in OPG(-/-) mice. Although OPG is normally expressed in arteries, OPG ligand (OPGL) and receptor activator of NF-kappaB (RANK) are not detected in the arterial walls of wild-type adult mice. Interestingly, OPGL and RANK transcripts are detected in the calcified arteries of OPG(-/-) mice. Furthermore, RANK transcript expression coincides with the presence of multinuclear osteoclast-like cells. These findings indicate that the OPG/OPGL/RANK signaling pathway may play an important role in both pathological and physiological calcification processes. Such findings may also explain the observed high clinical incidence of vascular calcification in the osteoporotic patient population.


Subject(s)
Bone Density/physiology , Calcinosis/physiopathology , Glycoproteins/metabolism , Osteoclasts/metabolism , Osteopetrosis/metabolism , Osteoporosis/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Acid Phosphatase/metabolism , Animals , Aorta/pathology , Blotting, Western , CHO Cells , Cathepsin K , Cathepsins/metabolism , Cricetinae , Femur/anatomy & histology , Femur/diagnostic imaging , Femur/metabolism , Glycoproteins/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Isoenzymes/metabolism , Mice , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoclasts/ultrastructure , Osteopetrosis/genetics , Osteoporosis/genetics , Osteoprotegerin , Radiography , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/metabolism , Tartrate-Resistant Acid Phosphatase
SELECTION OF CITATIONS
SEARCH DETAIL