Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Endocrinology ; 159(1): 260-271, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29069393

ABSTRACT

Sclerostin antibody (Scl-Ab) restored bone mass and strength in the ovariectomized rat model of postmenopausal osteoporosis. Increased bone mineral density (BMD) and decreased skeletal fragility fracture risk have been reported in postmenopausal osteoporotic women receiving Scl-Ab. In males, loss of androgen leads to rapid decreases in BMD and an increased risk of fragility fractures. We hypothesized that Scl-Ab could reverse the loss of bone mass and strength caused by androgen ablation in the orchiectomized (ORX) rat model of male osteoporosis. We treated 9-month-old ORX Sprague Dawley rats (3 months after ORX) subcutaneously twice weekly with vehicle or Scl-Ab (5 or 25 mg/kg) for 6 weeks (n = 10 per group). Both doses of Scl-Ab fully reversed the BMD deficit in the lumbar spine and femur and tibia in ORX rats. Microcomputed tomography showed that the bone mass in the fifth lumbar vertebral body, femur diaphysis, and femoral neck were dose-dependently restored by Scl-Ab. The bone strength at these sites increased significantly with Scl-Ab to levels matching those of sham-operated controls and correlated positively with improvements in bone mineral content, demonstrating bone quality maintenance. Dynamic histomorphometry of the tibial diaphysis and second lumbar vertebral body demonstrated that Scl-Ab significantly increased bone formation on periosteal, endocortical, and trabecular surfaces and significantly decreased bone resorption on endocortical and trabecular surfaces. The effects of Scl-Ab on increasing bone formation and decreasing bone resorption led to restoration of bone mass and strength in androgen-deficient rats. These findings support the ongoing evaluation of Scl-Ab as a potential therapeutic agent for osteoporosis in men.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Density Conservation Agents/therapeutic use , Bone Morphogenetic Proteins/immunology , Bone and Bones/drug effects , Disease Models, Animal , Genetic Markers/immunology , Osteogenesis/drug effects , Osteoporosis/drug therapy , Absorptiometry, Photon , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Bone Density/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/adverse effects , Bone Morphogenetic Proteins/metabolism , Bone Resorption/prevention & control , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Dose-Response Relationship, Drug , Injections, Subcutaneous , Male , Molecular Targeted Therapy , Orchiectomy , Organ Size/drug effects , Osteoporosis/diagnostic imaging , Osteoporosis/pathology , Prostate/drug effects , Prostate/pathology , Rats, Sprague-Dawley , Seminal Vesicles/drug effects , Seminal Vesicles/pathology , Shear Strength/drug effects , Weight Gain/drug effects , X-Ray Microtomography
2.
Bone ; 105: 163-172, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28867373

ABSTRACT

Sustained elevation of parathyroid hormone (PTH) is catabolic to cortical bone, as evidenced by deterioration in bone structure (cortical porosity), and is a major factor for increased fracture risk in chronic kidney disease (CKD). Etelcalcetide (AMG 416), a novel peptide agonist of the calcium-sensing receptor, reduces PTH levels in subtotal nephrectomized (Nx) rats and in hemodialysis patients with secondary hyperparathyroidism (SHPT) in clinical studies; however, effects of etelcalcetide on bone have not been determined. In a rat model of established SHPT with renal osteodystrophy, etelcalcetide or vehicle was administered by subcutaneous (s.c.) injection to subtotal Nx rats with elevated PTH (>750pg/mL) once per day for 6weeks. Sham-operated rats receiving vehicle (s.c.) served as non-SHPT controls. Prior to treatment, significant increases in serum creatinine (2-fold), blood urea nitrogen (BUN, 3-fold), PTH (5-fold), fibroblast growth factor-23 (FGF23; 13-fold) and osteocalcin (12-fold) were observed in SHPT rats compared to non-SHPT controls. Elevations in serum creatinine and BUN were unaffected by treatment with vehicle or etelcalcetide. In contrast, etelcalcetide significantly decreased PTH, FGF23 and osteocalcin, whereas vehicle treatment did not. Cortical bone porosity increased and bone strength decreased in vehicle-treated SHPT rats compared to non-SHPT controls. Cortical bone structure improved and energy to failure was significantly greater in SHPT rats treated with etelcalcetide compared to vehicle. Mineralization lag time and marrow fibrosis were significantly reduced by etelcalcetide. In conclusion, etelcalcetide reduced bone turnover, attenuated mineralization defect and marrow fibrosis, and preserved cortical bone structure and bone strength by lowering PTH in subtotal Nx rats with established SHPT.


Subject(s)
Cortical Bone/physiopathology , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/physiopathology , Nephrectomy , Peptides/therapeutic use , Receptors, Calcium-Sensing/agonists , Animals , Biomechanical Phenomena/drug effects , Blood Urea Nitrogen , Calcium/blood , Cortical Bone/drug effects , Creatinine/blood , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Hyperparathyroidism, Secondary/blood , Hyperplasia , Kidney Function Tests , Male , Osteocalcin/blood , Parathyroid Glands/pathology , Parathyroid Hormone/blood , Peptides/pharmacology , Phosphorus/blood , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase/blood
3.
J Bone Miner Res ; 32(4): 834-845, 2017 04.
Article in English | MEDLINE | ID: mdl-27505721

ABSTRACT

Results of prior studies suggest that fibroblast growth factor 21 (FGF21) may be involved in bone turnover and in the actions of peroxisome proliferator-activated receptor (PPAR) α and γ in mice. We have conducted independent studies to examine the effects of FGF21 on bone homeostasis and the role of FGF21 in PPARα and γ actions. High-fat-diet-induced obesity (DIO) mice were administered vehicle or recombinant human FGF21 (rhFGF21) intraperitoneally at 0 (vehicle), 0.1, 1, and 3 mg/kg daily for 2 weeks. Additional groups of DIO mice received water or 10 mg/kg rosiglitazone daily. Mice treated with rhFGF21 or rosiglitazone showed expected metabolic improvements in glucose, insulin, and lipid levels. However, bone loss was not detected in rhFGF21-treated mice by dual-energy X-ray absorptiometry (DXA), micro-CT, and histomorphometric analyses. Mineral apposition rate, a key bone formation parameter, was unchanged by rhFGF21, while significantly decreased by rosiglitazone in DIO mice. Bone resorption markers, OPG/RANKL mRNA expression, and histological bone resorption indices were unchanged by rhFGF21 or rosiglitazone. Bone marrow fat was unchanged by rhFGF21, while increased by rosiglitazone. Furthermore, FGF21 knockout mice did not show high bone mass phenotype. Treatment with PPARα or PPARγ agonists caused similar metabolic effects in FGF21 knockout and wild-type mice. These results contrast with previous findings and suggest that FGF21 is not critical for bone homeostasis or actions of PPARα and PPARγ. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Density , Fibroblast Growth Factors , Gene Expression Regulation/drug effects , Homeostasis , PPAR alpha , PPAR gamma , Animals , Bone Density/drug effects , Bone Density/genetics , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Glucose/metabolism , Homeostasis/drug effects , Homeostasis/genetics , Humans , Insulin/genetics , Insulin/metabolism , Male , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/metabolism , Osteoprotegerin/biosynthesis , Osteoprotegerin/genetics , PPAR alpha/agonists , PPAR alpha/biosynthesis , PPAR alpha/genetics , PPAR gamma/agonists , PPAR gamma/biosynthesis , PPAR gamma/genetics , RANK Ligand/biosynthesis , RANK Ligand/genetics , Rosiglitazone , Thiazolidinediones/pharmacology
4.
Bone ; 50(3): 628-37, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22154841

ABSTRACT

We examined age-related changes in biochemical markers and regulators of osteoblast and osteoclast activity in C57BL/6 mice to assess their utility in explaining age-related changes in bone. Several recently discovered regulators of osteoclasts and osteoblasts were also measured to assess concordance between their systemic levels versus their levels in marrow plasma, to which bone cells are directly exposed. MicroCT of 6-, 12-, and 24-month-old mice indicated an early age-related loss of trabecular bone volume and surface, followed by endocortical bone loss and periosteal expansion. Trabecular bone loss temporally correlated with reductions in biomarkers of bone formation and resorption in both peripheral blood and bone marrow. Endocortical bone loss and periosteal bone gain were not reflected in these protein biomarkers, but were well correlated with increased expression of osteocalcin, rank, tracp5b, and cathepsinK in RNA extracted from cortical bone. While age-related changes in bone turnover markers remained concordant in blood versus marrow, aging led to divergent changes in blood versus marrow for the bone cell regulators RANKL, OPG, sclerostin, DKK1, and serotonin. Bone expression of runx2 and osterix increased progressively with aging and was associated with an increase in the number of osteoprogenitors and osteoclast precursors. In summary, levels of biochemical markers of bone turnover in blood and bone marrow plasma were predictive of an age-related loss of trabecular surfaces in adult C57BL/6 mice, but did not predict gains in cortical surfaces resulting from cortical expansion. Unlike these turnover markers, a panel of bone cell regulatory proteins exhibited divergent age-related changes in marrow versus peripheral blood, suggesting that their circulating levels may not reflect local levels to which osteoclasts and osteoblasts are directly exposed.


Subject(s)
Aging/metabolism , Bone Remodeling/physiology , Bone and Bones/metabolism , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Animals , Biomarkers/blood , Bone Marrow/metabolism , Cells, Cultured , Male , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism
5.
J Bone Miner Res ; 26(11): 2610-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21773994

ABSTRACT

The physiological role of Dickkopf-1 (Dkk1) during postnatal bone growth in rodents and in adult rodents was examined utilizing an antibody to Dkk1 (Dkk1-Ab) that blocked Dkk1 binding to both low density lipoprotein receptor-related protein 6 (LRP6) and Kremen2, thereby preventing the Wnt inhibitory activity of Dkk1. Treatment of growing mice and rats with Dkk1-Ab resulted in a significant increase in bone mineral density because of increased bone formation. In contrast, treatment of adult ovariectomized rats did not appreciably impact bone, an effect that was associated with decreased Dkk1 expression in the serum and bone of older rats. Finally, we showed that Dkk1 plays a prominent role in adult bone by mediating fracture healing in adult rodents. These data suggest that, whereas Dkk1 significantly regulates bone formation in younger animals, its role in older animals is limited to pathologies that lead to the induction of Dkk1 expression in bone and/or serum, such as traumatic injury.


Subject(s)
Aging/metabolism , Bone and Bones/injuries , Bone and Bones/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Osteogenesis/physiology , Aging/drug effects , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/pharmacology , Bone Density/drug effects , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/physiopathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cell Line , Estrogens/deficiency , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Fracture Healing/drug effects , Humans , Intercellular Signaling Peptides and Proteins/blood , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Male , Mice , Osteogenesis/drug effects , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects , X-Ray Microtomography
6.
Ann Rheum Dis ; 69(12): 2152-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20858621

ABSTRACT

UNLABELLED: Introduction Inflammation is a major risk factor for systemic bone loss. Proinflammatory cytokines like tumour necrosis factor (TNF) affect bone homeostasis and induce bone loss. It was hypothesised that impaired bone formation is a key component in inflammatory bone loss and that Dkk-1, a Wnt antagonist, is a strong inhibitor of osteoblast-mediated bone formation. METHODS: TNF transgenic (hTNFtg) mice were treated with neutralising antibodies against TNF, Dkk-1 or a combination of both agents. Systemic bone architecture was analysed by bone histomorphometry. The expression of ß-catenin, osteoprotegerin and osteocalcin was analysed. In vitro, primary osteoblasts were stimulated with TNF and analysed for their metabolic activity and expression of Dkk-1 and sclerostin. Sclerostin expression and osteocyte death upon Dkk-1 blockade were analysed in vivo. RESULTS: Neutralisation of Dkk-1 completely protected hTNFtg mice from inflammatory bone loss by preventing TNF-mediated impaired osteoblast function and enhanced osteoclast activity. These findings were accompanied by enhanced skeletal expression of ß-catenin, osteocalcin and osteoprotegerin. In vitro, TNF rapidly increased Dkk-1 expression in primary osteoblasts and effectively blocked osteoblast differentiation. Moreover, blockade of Dkk-1 not only rescued impaired osteoblastogenesis but also neutralised TNF-mediated sclerostin expression in fully differentiated osteoblasts in vitro and in vivo. CONCLUSIONS: These findings indicate that low bone formation and expression of Dkk-1 trigger inflammatory bone loss. Dkk-1 blocks osteoblast differentiation, induces sclerostin expression and leads to osteocyte death. Inhibition of Dkk-1 may thus be considered as a potent strategy to protect bone from inflammatory damage.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Bone Morphogenetic Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/metabolism , Bone Resorption/prevention & control , Cell Differentiation/drug effects , Cells, Cultured , Genetic Markers , Glycoproteins , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Osteoblasts/drug effects , Osteoblasts/physiology , Osteocytes/metabolism , Osteogenesis/drug effects , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
7.
Bone ; 45(4): 669-76, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19539794

ABSTRACT

Orchiectomized (ORX) rats were used to examine the extent to which their increased bone resorption and decreased bone density might relate to increases in RANKL, an essential cytokine for bone resorption. Serum testosterone declined by >95% in ORX rats 1 and 2 weeks after surgery (p<0.05 versus sham controls), with no observed changes in serum RANKL. In contrast, RANKL in bone marrow plasma and bone marrow cell extracts was significantly increased (by approximately 100%) 1 and 2 weeks after ORX. Regression analyses of ORX and sham controls revealed a significant inverse correlation between testosterone and RANKL levels measured in marrow cell extracts (R=-0.58), while marrow plasma RANKL correlated positively with marrow plasma TRACP-5b, an osteoclast marker (R=0.63). The effects of RANKL inhibition were then studied by treating ORX rats for 6 weeks with OPG-Fc (10 mg/kg, twice/week SC) or with PBS, beginning immediately after surgery. Sham controls were treated with PBS. Vehicle-treated ORX rats showed significant deficits in BMD of the femur/tibia and lower trabecular bone volume in the distal femur (p<0.05 versus sham). OPG-Fc treatment of ORX rats increased femur/tibia BMD and trabecular bone volume to levels that significantly exceeded values for ORX or sham controls. OPG-Fc reduced trabecular osteoclast surfaces in ORX rats by 99%, and OPG-Fc also prevented ORX-related increases in endocortical eroded surface and ORX-related reductions in periosteal bone formation rate. Micro-CT of lumbar vertebrae from OPG-Fc-treated ORX rats demonstrated significantly greater cortical and trabecular bone volume and density versus ORX-vehicle controls. In summary, ORX rats exhibited increased RANKL protein in bone marrow plasma and in bone marrow cells, with no changes in serum RANKL. Data from regression analyses were consistent with a potential role for testosterone in suppressing RANKL production in bone marrow, and also suggested that soluble RANKL in bone marrow might promote bone resorption. RANKL inhibition prevented ORX-related deficits in trabecular BMD, trabecular architecture, and periosteal bone formation while increasing cortical and trabecular bone volume and density. These results support the investigation of RANKL inhibition as a strategy for preventing bone loss associated with androgen ablation or deficiency.


Subject(s)
Bone Marrow/metabolism , Bone Resorption/prevention & control , Orchiectomy , Osteoprotegerin/metabolism , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Acid Phosphatase/blood , Animals , Bone Density/drug effects , Bone Marrow/drug effects , Bone Resorption/blood , Femur Neck/diagnostic imaging , Femur Neck/drug effects , Femur Neck/pathology , Humans , Isoenzymes/blood , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Male , Osteoprotegerin/pharmacology , Rats , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase , X-Ray Microtomography
8.
Am J Pathol ; 174(6): 2160-71, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19389927

ABSTRACT

Intracortical porosities and marrow fibrosis are hallmarks of hyperparathyroidism and are present in bones of transgenic mice expressing constitutively active parathyroid hormone/parathyroid hormone-related protein receptors (PPR*Tg). Cortical porosity is the result of osteoclast activity; however, the etiology of marrow fibrosis is poorly understood. While osteoclast numbers and activity are regulated by osteoprotegerin (OPG), bisphosphonates suppress osteoclast activity but not osteoclast numbers. We therefore used OPG and bisphosphonates to evaluate the extent to which osteoclasts, as opposed to bone resorption, regulate marrow fibrosis in PPR*Tg mice after treatment of animals with vehicle, OPG, alendronate, or zoledronate. All three agents similarly increased trabecular bone volume in both PPR*Tg and control mice, suggesting that trabecular bone resorption was comparably suppressed by these agents. However, the number of trabecular osteoclasts was greatly decreased by OPG but not by either alendronate or zoledronate. Furthermore, intracortical porosity and marrow fibrosis were virtually abolished by OPG treatment, whereas alendronate and zoledronate only partially reduced these two parameters. The greater reductions in cortical porosity and increments in cortical bone mineral density with OPG in PPR*Tg mice were associated with greater improvements in bone strength. The differential effect of OPG versus bisphosphonates on marrow fibrosis, despite similar effects on trabecular bone volume, suggests that marrow fibrosis was related not only to bone resorption but also to the presence of osteoclasts.


Subject(s)
Bone and Bones/metabolism , Osteoprotegerin/metabolism , Primary Myelofibrosis/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Alendronate/pharmacology , Animals , Biomechanical Phenomena , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/pathology , Diphosphonates/pharmacology , Disease Models, Animal , Humans , Hyperparathyroidism/metabolism , Hyperparathyroidism/pathology , Imidazoles/pharmacology , Immunohistochemistry , Male , Mice , Mice, Transgenic , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/pharmacology , Porosity , Primary Myelofibrosis/pathology , Receptor, Parathyroid Hormone, Type 1/genetics , Tomography, X-Ray Computed , Zoledronic Acid
9.
J Bone Miner Res ; 24(2): 182-95, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19016581

ABSTRACT

RANKL is a TNF family member that mediates osteoclast formation, activation, and survival by activating RANK. The proresorptive effects of RANKL are prevented by binding to its soluble inhibitor osteoprotegerin (OPG). Recombinant human OPG-Fc recognizes RANKL from multiple species and reduced bone resorption and increased bone volume, density, and strength in a number of rodent models of bone disease. The clinical development of OPG-Fc was discontinued in favor of denosumab, a fully human monoclonal antibody that specifically inhibits primate RANKL. Direct binding assays showed that denosumab bound to human RANKL but not to murine RANKL, human TRAIL, or other human TNF family members. Denosumab did not suppress bone resorption in normal mice or rats but did prevent the resorptive response in mice challenged with a human RANKL fragment encoded primarily by the fifth exon of the RANKL gene. To create mice that were responsive to denosumab, knock-in technology was used to replace exon 5 from murine RANKL with its human ortholog. The resulting "huRANKL" mice exclusively express chimeric (human/murine) RANKL that was measurable with a human RANKL assay and that maintained bone resorption at slightly reduced levels versus wildtype controls. In young huRANKL mice, denosumab and OPG-Fc each reduced trabecular osteoclast surfaces by 95% and increased bone density and volume. In adult huRANKL mice, denosumab reduced bone resorption, increased cortical and cancellous bone mass, and improved trabecular microarchitecture. These huRANKL mice have potential utility for characterizing the activity of denosumab in a variety of murine bone disease models.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Density/drug effects , Bone Resorption/drug therapy , Bone Resorption/physiopathology , Gene Knock-In Techniques , RANK Ligand/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibody Affinity/drug effects , Antibody Specificity/drug effects , Bone and Bones/drug effects , Bone and Bones/pathology , Denosumab , Humans , Hypercalcemia/drug therapy , Mice , Molecular Sequence Data , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoprotegerin/metabolism , Phenotype , Protein Binding/drug effects , RANK Ligand/chemistry , RANK Ligand/genetics , RANK Ligand/pharmacokinetics , RANK Ligand/pharmacology , RANK Ligand/therapeutic use , X-Ray Microtomography
10.
J Bone Miner Res ; 23(5): 672-82, 2008 May.
Article in English | MEDLINE | ID: mdl-18433301

ABSTRACT

INTRODUCTION: Ovariectomy (OVX) results in bone loss caused by increased bone resorption. RANKL is an essential mediator of bone resorption. We examined whether the RANKL inhibitor osteoprotegerin (OPG) would preserve bone volume, density, and strength in OVX rats. MATERIALS AND METHODS: Rats were OVX or sham-operated at 3 mo of age. Sham controls were treated for 6 wk with vehicle (Veh, PBS). OVX rats were treated with Veh or human OPG-Fc (10 mg/kg, 2/wk). Serum RANKL and TRACP5b was measured by ELISA. BMD of lumbar vertebrae (L(1)-L(5)) and distal femur was measured by DXA. Right distal femurs were processed for bone histomorphometry. Left femurs and the fifth lumbar vertebra (L(5)) were analyzed by muCT and biomechanical testing, and L(6) was analyzed for ash weight. RESULTS: OVX was associated with significantly greater serum RANKL and osteoclast surface and with reduced areal and volumetric BMD. OPG markedly reduced osteoclast surface and serum TRACP5b while completely preventing OVX-associated bone loss in the lumbar vertebrae, distal femur, and femur neck. Vertebrae from OPG-treated rats had increased dry and ash weight, with no significant differences in tissue mineralization versus OVX controls. muCT showed that trabecular compartments in OVX-OPG rats had significantly greater bone volume fraction, vBMD, bone area, trabecular thickness, and number, whereas their cortical compartments had significantly greater bone area (p < 0.05 versus OVX-Veh). OPG improved cortical area in L(5) and the femur neck to levels that were significantly greater than OVX or sham controls (p < 0.05). Biomechanical testing of L(5) and femur necks showed significantly greater maximum load values in the OVX-OPG group (p < 0.05 versus OVX-Veh). Bone strength at both sites was linearly correlated with total bone area (r(2) = 0.54-0.74, p < 0.0001), which was also significantly increased by OPG (p < 0.05 versus OVX). CONCLUSIONS: OPG treatment prevented bone loss, preserved trabecular architecture, and increased cortical area and bone strength in OVX rats.


Subject(s)
Bone and Bones/anatomy & histology , Osteoprotegerin/physiology , Ovariectomy , RANK Ligand/antagonists & inhibitors , Animals , Female , RANK Ligand/genetics , Rats , Rats, Sprague-Dawley
11.
J Bone Miner Res ; 23(6): 860-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18269310

ABSTRACT

INTRODUCTION: Sclerosteosis is a rare high bone mass genetic disorder in humans caused by inactivating mutations in SOST, the gene encoding sclerostin. Based on these data, sclerostin has emerged as a key negative regulator of bone mass. We generated SOST knockout (KO) mice to gain a more detailed understanding of the effects of sclerostin deficiency on bone. MATERIALS AND METHODS: Gene targeting was used to inactivate SOST and generate a line of SOST KO mice. Radiography, densitometry, microCT, histomorphometry, and mechanical testing were used to characterize the impact of sclerostin deficiency on bone in male and female mice. Comparisons were made between same sex KO and wildtype (WT) mice. RESULTS: The results for male and female SOST KO mice were similar, with differences only in the magnitude of some effects. SOST KO mice had increased radiodensity throughout the skeleton, with general skeletal morphology being normal in appearance. DXA analysis of lumbar vertebrae and whole leg showed that there was a significant increase in BMD (>50%) at both sites. microCT analysis of femur showed that bone volume was significantly increased in both the trabecular and cortical compartments. Histomorphometry of trabecular bone revealed a significant increase in osteoblast surface and no significant change in osteoclast surface in SOST KO mice. The bone formation rate in SOST KO mice was significantly increased for trabecular bone (>9-fold) at the distal femur, as well as for the endocortical and periosteal surfaces of the femur midshaft. Mechanical testing of lumbar vertebrae and femur showed that bone strength was significantly increased at both sites in SOST KO mice. CONCLUSIONS: SOST KO mice have a high bone mass phenotype characterized by marked increases in BMD, bone volume, bone formation, and bone strength. These results show that sclerostin is a key negative regulator of a powerful, evolutionarily conserved bone formation pathway that acts on both trabecular and cortical bone.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone and Bones/metabolism , Gene Deletion , Osteogenesis , Adaptor Proteins, Signal Transducing , Animals , Biomarkers/blood , Bone Density , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Bone and Bones/diagnostic imaging , Calcium/blood , Female , Genetic Markers/genetics , Glycoproteins , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Knockout , Phenotype , Phosphates/blood , Stress, Mechanical , Tomography, X-Ray Computed
12.
Bone ; 39(4): 754-66, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16730481

ABSTRACT

Mutations affecting the activity of the Wnt co-receptors LRP5 and LRP6 that cause alterations in skeletal biology confirmed the involvement of Wnt signaling in bone formation. We evaluated the potential role of Dkk1, an inhibitor of LRP5/6 activity, in bone formation by examining the normal expression pattern of Dkk1 in normal young mice and by assessing the consequences of osteoblast overexpression of Dkk1 in transgenic mice. Endogenous Dkk1 expression was detected primarily in osteoblasts and osteocytes. Transgenic over-expression of Dkk1 using two different rat collagen 1A1 promoters resulted in distinct bone phenotypes. More widespread Dkk1 expression (driven by the Col1A1 3.6 kb promoter) yielded osteopenia with forelimb deformities and hairlessness, while expression restricted to osteoblasts (driven by the Col1A1 2.3 kb promoter) induced severe osteopenia without limb defects or alopecia. The decrease in bone mass in vivo resulted from a significant 49% reduction in osteoblast numbers and was reflected in a 45% reduction in serum osteocalcin concentration; an in vitro study revealed that Dkk1 caused a dose-dependent suppression of osteoblast matrix mineralization. These data indicate that Dkk1 may directly influence bone formation and suggest that osteopenia develops in mice over-expressing Dkk1 at least in part due to diminished bone formation resulting from reduced osteoblast numbers.


Subject(s)
Bone Diseases, Metabolic/physiopathology , Bone and Bones/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Wnt Proteins/physiology , 3T3 Cells , Animals , Bone Density , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Bone and Bones/pathology , Bone and Bones/physiopathology , Cells, Cultured , Embryo, Mammalian/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , LDL-Receptor Related Proteins/metabolism , Male , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/blood , Osteogenesis/genetics , Osteogenesis/physiology , Pregnancy , Rats , Recombinant Proteins/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism
13.
J Bone Miner Res ; 20(10): 1756-65, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16160733

ABSTRACT

UNLABELLED: RANKL is an essential mediator of bone erosions, but the role of RANKL in systemic bone loss had not been studied in arthritis. RANKL protein was increased in rat joint extracts and serum at the earliest stages of arthritis. Osteoprotegerin (OPG) treatment reversed local and systemic bone loss, suggesting that RANKL is both a marker and mediator of bone loss in arthritis. INTRODUCTION: RANKL is well established as an essential mediator of bone erosions in inflammatory arthritis, but the role of RANKL in systemic bone loss in arthritis had not been studied. We hypothesized that serum RANKL could serve as both a mediator and as a novel biomarker for local and systemic bone loss in arthritis. We challenged this hypothesis in two established rat models of inflammatory arthritis. We sought to determine whether serum RANKL was elevated early in disease progression and whether RANKL suppression could prevent both local and systemic bone loss in these models. MATERIALS AND METHODS: Detailed time-course studies were conducted in animals with collagen-induced (CIA) or adjuvant-induced (AIA) arthritis to evaluate the onset and progression of inflammation (paw swelling), bone erosions, osteoclast numbers, and RANKL protein levels in arthritic joints and in serum. Additional CIA and AIA rats (n=8/group) received placebo (PBS) or recombinant OPG (3 mg/kg three times weekly) for 10 days beginning 4 days after disease onset (first macroscopic evidence of hind paw erythema and edema) to assess the role of RANKL in local and systemic bone loss. RESULTS: RANKL protein was significantly elevated in the joints and serum of CIA and AIA rats within 1-2 days of disease onset. Increased RANKL levels were associated with local (hind paw) and systemic (vertebral) osteopenia in both models. The RANKL inhibitor OPG prevented local and systemic osteopenia in both models of established disease. CONCLUSIONS: RANKL protein is significantly increased both locally and systemically during the earliest stages of inflammatory arthritis in rats, suggesting that serum RANKL might have prognostic value for bone erosions and systemic osteopenia in this condition. RANKL inhibition through OPG prevented local and systemic bone loss in these arthritis models, suggesting that RANKL inhibition is a promising new approach for treating bone loss in arthritis.


Subject(s)
Arthritis, Experimental/blood , Bone Diseases, Metabolic/blood , Carrier Proteins/blood , Membrane Glycoproteins/blood , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/drug therapy , Biomarkers/blood , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/etiology , Disease Models, Animal , Female , Glycoproteins/administration & dosage , Inflammation/blood , Inflammation/complications , Inflammation/drug therapy , Joints/metabolism , Male , Osteoprotegerin , RANK Ligand , Rats , Rats, Inbred Lew , Receptors, Cytoplasmic and Nuclear/administration & dosage , Receptors, Tumor Necrosis Factor/administration & dosage
14.
Endocrinology ; 146(8): 3235-43, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15845617

ABSTRACT

Humoral hypercalcemia of malignancy (HHM) is mediated primarily by skeletal and renal responses to tumor-derived PTHrP. PTHrP mobilizes calcium from bone by inducing the expression of receptor activator for nuclear factor-kappaB ligand (RANKL), a protein that is essential for osteoclast formation, activation, and survival. RANKL does not influence renal calcium reabsorption, so RANKL inhibition is a rational approach to selectively block, and thereby reveal, the relative contribution of bone calcium to HHM. We used the RANKL inhibitor osteoprotegerin (OPG) to evaluate the role of osteoclast-mediated hypercalcemia in two murine models of HHM. Hypercalcemia was induced either by sc inoculation of syngeneic colon (C-26) adenocarcinoma cells or by sc injection of high-dose recombinant PTHrP (0.5 mg/kg, s.c., twice per day). In both models, OPG (0.2-5 mg/kg) caused rapid reversal of established hypercalcemia, and the speed and duration of hypercalcemia suppression were significantly greater with OPG (5 mg/kg) than with high-dose bisphosphonates (pamidronate or zoledronic acid, 5 mg/kg). OPG also caused greater reductions in osteoclast surface and biochemical markers of bone resorption compared with either bisphosphonate. In both models, hypercalcemia gradually returned despite clear evidence of ongoing suppression of bone resorption by OPG. These data demonstrate that osteoclasts and RANKL are important mediators of HHM, particularly in the early stages of the condition. Aggressive antiresorptive therapy with a RANKL inhibitor therefore might be a rational approach to controlling HHM.


Subject(s)
Adenocarcinoma/prevention & control , Bone Resorption/prevention & control , Carrier Proteins/antagonists & inhibitors , Colonic Neoplasms/prevention & control , Diphosphonates/pharmacology , Glycoproteins/pharmacology , Hypercalcemia/prevention & control , Membrane Glycoproteins/antagonists & inhibitors , Adenocarcinoma/blood , Animals , Antineoplastic Agents/pharmacology , Calcium/blood , Cell Line, Tumor , Colonic Neoplasms/blood , Disease Models, Animal , Humans , Hypercalcemia/etiology , Ligands , Mice , NF-kappa B/metabolism , Osteoprotegerin , Pamidronate , Parathyroid Hormone-Related Protein/physiology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor
15.
Toxicol Pathol ; 32(3): 275-94, 2004.
Article in English | MEDLINE | ID: mdl-15204970

ABSTRACT

Artemin (ART) signals through the GFR alpha-3/RET receptor complex to support sympathetic neuron development. Here we show that ART also influences autonomic elements in adrenal medulla and enteric and pelvic ganglia. Transgenic mice over-expressing Art throughout development exhibited systemic autonomic neural lesions including fusion of adrenal medullae with adjacent paraganglia, adrenal medullary dysplasia, and marked enlargement of sympathetic (superior cervical and sympathetic chain ganglia) and parasympathetic (enteric, pelvic) ganglia. Changes began by gestational day 12.5 and formed progressively larger masses during adulthood. Art supplementation in wild type adult mice by administering recombinant protein or an Art-bearing retroviral vector resulted in hyperplasia or neuronal metaplasia at the adrenal corticomedullary junction. Expression data revealed that Gfr alpha-3 is expressed during development in the adrenal medulla, sensory and autonomic ganglia and their projections, while Art is found in contiguous mesenchymal domains (especially skeleton) and in certain nerves. Intrathecal Art therapy did not reduce hypalgesia in rats following nerve ligation. These data (1) confirm that ART acts as a differentiation factor for autonomic (chiefly sympathoadrenal but also parasympathetic) neurons, (2) suggest a role for ART overexpression in the genesis of pheochromocytomas and paragangliomas, and (3) indicate that ART is not a suitable therapy for peripheral neuropathy.


Subject(s)
Autonomic Nervous System/embryology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Peripheral Nerves/embryology , Adult , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiology , Autonomic Nervous System Diseases/pathology , Blotting, Southern , Cells, Cultured , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peripheral Nerves/drug effects , Peripheral Nerves/physiology , Peripheral Nervous System Diseases/pathology , Polymerase Chain Reaction , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL