Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Bone Joint Surg Am ; 105(15): 1145-1155, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37159527

ABSTRACT

BACKGROUND: Fracture repair involves the reactivation of developmental signaling cascades, including Wnt signaling that stimulates bone formation and bone regeneration. Rodent data indicate that dual inhibition of the Wnt signaling antagonists sclerostin and Dickkopf-1 (DKK1) increases callus bone volume and strength while increasing bone mass systemically. METHODS: We evaluated the effects of 16 weeks of subcutaneously administered carrier solution (vehicle, VEH), anti-sclerostin antibody (Scl-Ab), anti-DKK1 antibody (DKK1-Ab), or Scl-Ab plus DKK1-Ab combination therapy (COMBO) on ulnar osteotomy healing in nonhuman primates (cynomolgus monkeys; 20 to 22 per group). RESULTS: Scl-Ab and COMBO therapy increased systemic markers of bone formation versus VEH, with COMBO leading to synergistic increases versus Scl-Ab or DKK1-Ab monotherapies. The COMBO and Scl-Ab groups showed reduced serum markers of bone resorption versus VEH. The COMBO and DKK1-Ab groups exhibited greater callus bone mineral density (BMD), torsional stiffness, and torsional rigidity versus VEH. Lumbar vertebrae from the Scl-Ab and COMBO groups showed greater BMD and bone formation rate versus VEH, and the femoral mid-diaphysis of the Scl-Ab and COMBO groups showed greater periosteal and endocortical bone formation rates versus VEH. CONCLUSIONS: DKK1-Ab increased BMD and strength at the ulnar osteotomy site, Scl-Ab increased bone formation and BMD at uninjured skeletal sites, and Scl-Ab plus DKK1-Ab combination therapy induced all of these effects, in some cases to a greater degree versus 1 or both monotherapies. These results in nonhuman primates suggest that DKK1 preferentially regulates bone healing while sclerostin preferentially regulates systemic bone mass. CLINICAL RELEVANCE: Combination therapy with antibodies against sclerostin and DKK1 may offer a promising therapeutic strategy for both fracture treatment and fracture prevention.


Subject(s)
Fracture Healing , Fractures, Bone , Animals , Antibodies, Monoclonal/therapeutic use , Bone and Bones , Bone Density , Osteogenesis/physiology , Primates
2.
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
3.
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
4.
Nat Commun ; 7: 11505, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27230681

ABSTRACT

Inhibition of the Wnt antagonist sclerostin increases bone mass in patients with osteoporosis and in preclinical animal models. Here we show increased levels of the Wnt antagonist Dickkopf-1 (DKK-1) in animals treated with sclerostin antibody, suggesting a negative feedback mechanism that limits Wnt-driven bone formation. To test our hypothesis that co-inhibition of both factors further increases bone mass, we engineer a first-in-class bispecific antibody with single residue pair mutations in the Fab region to promote efficient and stable cognate light-heavy chain pairing. We demonstrate that dual inhibition of sclerostin and DKK-1 leads to synergistic bone formation in rodents and non-human primates. Furthermore, by targeting distinct facets of fracture healing, the bispecific antibody shows superior bone repair activity compared with monotherapies. This work supports the potential of this agent both for treatment and prevention of fractures and offers a promising therapeutic approach to reduce the burden of low bone mass disorders.


Subject(s)
Antibodies, Bispecific/administration & dosage , Fractures, Bone/drug therapy , Fractures, Bone/physiopathology , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Density , Disease Models, Animal , Female , Fractures, Bone/genetics , Fractures, Bone/metabolism , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Macaca fascicularis , Male , Mice , Mice, Knockout , Osteogenesis/drug effects , Rats , Rats, Sprague-Dawley , Wnt Signaling Pathway/drug effects , Wound Healing/drug effects
5.
Endocrinology ; 155(12): 4785-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25259718

ABSTRACT

The effects of up to 26 weeks of sclerostin antibody (Scl-Ab) treatment were investigated in ovariectomized (OVX) rats. Two months after surgery, 6-month-old osteopenic OVX rats were treated with vehicle or Scl-Ab (25 mg/kg, sc, one time per week) for 6, 12, or 26 weeks. In vivo dual-energy x-ray absorptiometry analysis demonstrated that the bone mineral density of lumbar vertebrae and femur-tibia increased progressively through 26 weeks of Scl-Ab treatment along with progressive increases in trabecular and cortical bone mass and bone strength at multiple sites. There was a strong correlation between bone mass and maximum load at lumbar vertebra, femoral neck, and diaphysis at weeks 6 and 26. Dynamic histomorphometric analysis showed that lumbar trabecular and tibial shaft endocortical and periosteal bone formation rates (BFR/BS) increased and peaked at week 6 with Scl-Ab-treatment; thereafter trabecular and endocortical BFR/BS gradually declined but remained significantly greater than OVX controls at week 26, whereas periosteal BFR/BS returned to OVX control levels at week 26. In the tibia metaphysis, trabecular BFR/BS in the Scl-Ab treated group remained elevated from week 6 to week 26. The osteoclast surface and eroded surface were significantly lower in Scl-Ab-treated rats than in OVX controls at all times. In summary, bone mass and strength increased progressively over 26 weeks of Scl-Ab treatment in adult OVX rats. The early gains were accompanied by increased cortical and trabecular bone formation and reduced osteoclast activity, whereas later gains were attributed to residual endocortical and trabecular osteoblast stimulation and persistently low osteoclast activity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone and Bones/drug effects , Osteoporosis/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Bone Density/drug effects , Bone Remodeling/drug effects , Drug Evaluation, Preclinical , Female , Genetic Markers , Ovariectomy , Random Allocation , Rats, Sprague-Dawley , X-Ray Microtomography
6.
Bone ; 67: 305-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25093263

ABSTRACT

Sclerostin (Scl) is an osteocyte protein that decreases bone formation, and its inhibition by neutralizing antibodies (Scl-Ab) increases bone formation, mass and strength. We investigated the effects of Scl-Ab in mature ovariectomized (OVX) rats with a mechanistic focus on longer-term responses of osteoclasts, osteoblasts and osteocytes. Four-month-old Sprague-Dawley rats had OVX or sham surgery. Two months later, sham controls received sc vehicle while OVX rats received vehicle (OVX-Veh) or Scl-Ab (25mg/kg) once weekly for 6 or 26weeks followed by necropsy (n=12/group). Terminal blood was collected for biochemistry, non-adherent marrow cells were harvested from femurs for ex vivo osteoclast formation assays, and vertebrae and tibiae were collected for dynamic histomorphometry and mRNA analyses. Scl-Ab treatment led to progressively thicker but fewer trabeculae in the vertebra, leading to increased trabecular bone volume and reduced trabecular surfaces. Scl-Ab also increased cortical bone volume in the tibia, via early periosteal expansion and progressive endocortical contraction. Scl-Ab significantly reduced parameters of bone resorption at week 6 relative to OVX-Veh controls, including reduced serum TRACP-5b, reduced capacity of marrow cells to form osteoclasts ex vivo, and >80% reductions in vertebral trabecular and tibial endocortical eroded surfaces. At week 26, serum TRACP-5b and ex vivo osteoclast formation were no longer reduced in the Scl-Ab group, but eroded surfaces remained >80% lower than in OVX-Veh controls without evidence for altered skeletal mRNA expression of opg or rankl. Scl-Ab significantly increased parameters of bone formation at week 6 relative to OVX-Veh controls, including increases in serum P1NP and osteocalcin, and increased trabecular, endocortical and periosteal bone formation rates (BFRs). At week 26, surface-referent trabecular BFR remained significantly increased in the Scl-Ab group versus OVX-Veh controls, but after adjusting for a reduced extent of trabecular surfaces, overall (referent-independent) trabecular BFR was no longer significantly elevated. Similarly, serum P1NP and osteocalcin were no longer significantly increased in the Scl-Ab group at week 26. Tibial endocortical and periosteal BFR were increased at week 6 in the Scl-Ab group versus OVX-Veh controls, while at week 26 only endocortical BFR remained increased. The Scl-Ab group exhibited significant increments in skeletal mRNA expression of several osteocyte genes, with sost showing the greatest induction in both the tibia and vertebra. We propose that Scl-Ab administration, and/or the gains in bone volume that result, may have increased osteocytic expression of Scl as a possible means of regulating gains in bone mass.


Subject(s)
Antibodies/pharmacology , Bone Morphogenetic Proteins/immunology , Genetic Markers/immunology , Animals , Antibodies/administration & dosage , Bone Density/drug effects , Female , Osteoclasts/drug effects , Osteogenesis/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley
7.
Endocrinology ; 152(9): 3312-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21733832

ABSTRACT

Clinical studies have revealed a blunting of the bone anabolic effects of parathyroid hormone treatment in osteoporotic patients in the setting of pre- or cotreatment with the antiresorptive agent alendronate (ALN). Sclerostin monoclonal antibody (Scl-Ab) is currently under clinical investigation as a new potential anabolic therapy for postmenopausal osteoporosis. The purpose of these experiments was to examine the influence of pretreatment or cotreatment with ALN on the bone anabolic actions of Scl-Ab in ovariectomized (OVX) rats. Ten-month-old osteopenic OVX rats were treated with ALN or vehicle for 6 wk, before the start of Scl-Ab treatment. ALN-pretreated OVX rats were switched to Scl-Ab alone or to a combination of ALN and Scl-Ab for another 6 wk. Vehicle-pretreated OVX rats were switched to Scl-Ab or continued on vehicle to serve as controls. Scl-Ab treatment increased areal bone mineral density, volumetric bone mineral density, trabecular and cortical bone mass, and bone strength similarly in OVX rats pretreated with ALN or vehicle. Serum osteocalcin and bone formation rate on trabecular, endocortical, and periosteal surfaces responded similarly to Scl-Ab in ALN or vehicle-pretreated OVX rats. Furthermore, cotreatment with ALN did not have significant effects on the increased bone formation, bone mass, and bone strength induced by Scl-Ab in the OVX rats that were pretreated with ALN. These results indicate that the increases in bone formation, bone mass, and bone strength with Scl-Ab treatment were not affected by pre- or cotreatment with ALN in OVX rats with established osteopenia.


Subject(s)
Alendronate/pharmacology , Antibodies, Monoclonal/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Density/drug effects , Bone Diseases, Metabolic/drug therapy , Bone Morphogenetic Proteins/immunology , Genetic Markers/immunology , Osteogenesis/drug effects , Acid Phosphatase/blood , Alendronate/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Bone Density/immunology , Bone Density Conservation Agents/therapeutic use , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/immunology , Bone and Bones/drug effects , Bone and Bones/immunology , Disease Models, Animal , Female , Isoenzymes/blood , Osteocalcin/blood , Osteogenesis/immunology , Ovariectomy , Rats , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase
8.
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
9.
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
10.
J Bone Miner Res ; 25(12): 2647-56, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20641040

ABSTRACT

The purpose of this study was to evaluate the effects of sclerostin inhibition by treatment with a sclerostin antibody (Scl-AbII) on bone formation, bone mass, and bone strength in an aged, gonad-intact male rat model. Sixteen-month-old male Sprague-Dawley rats were injected subcutaneously with vehicle or Scl-AbII at 5 or 25 mg/kg twice per week for 5 weeks (9-10/group). In vivo dual-energy X-ray absorptiometry (DXA) analysis showed that there was a marked increase in areal bone mineral density of the lumbar vertebrae (L(1) to L(5) ) and long bones (femur and tibia) in both the 5 and 25 mg/kg Scl-AbII-treated groups compared with baseline or vehicle controls at 3 and 5 weeks after treatment. Ex vivo micro-computed tomographic (µCT) analysis demonstrated improved trabecular and cortical architecture at the fifth lumbar vertebral body (L(5) ), femoral diaphysis (FD), and femoral neck (FN) in both Scl-AbII dose groups compared with vehicle controls. The increased cortical and trabecular bone mass was associated with a significantly higher maximal load of L(5) , FD, and FN in the high-dose group. Bone-formation parameters (ie, mineralizing surface, mineral apposition rate, and bone-formation rate) at the proximal tibial metaphysis and tibial shaft were markedly greater on trabecular, periosteal, and endocortical surfaces in both Scl-AbII dose groups compared with controls. These results indicate that sclerostin inhibition by treatment with a sclerostin antibody increased bone formation, bone mass, and bone strength in aged male rats and, furthermore, suggest that pharmacologic inhibition of sclerostin may represent a promising anabolic therapy for low bone mass in aged men.


Subject(s)
Aging/metabolism , Antibodies, Monoclonal/immunology , Bone Density/physiology , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone and Bones/anatomy & histology , Bone and Bones/metabolism , Osteogenesis , Absorptiometry, Photon , Animals , Bone Morphogenetic Proteins/metabolism , Bone and Bones/cytology , Bone and Bones/diagnostic imaging , Collagen Type I/metabolism , Genetic Markers , Male , Organ Size , Osteocalcin/blood , Rats , Rats, Sprague-Dawley , Serotonin/blood , Tomography, X-Ray Computed
11.
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
12.
J Bone Miner Res ; 24(9): 1552-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19419305

ABSTRACT

A large genome-wide, recessive, N-ethyl-N-nitrosourea (ENU)-induced mutagenesis screen was performed on a mixed C57BL/6J and C3H.SW-H2/SnJ mouse background to identify genes regulating bone mass. Approximately 6500 male and female G(3) hybrid mice were phenotyped at 8 and 10 wk of age by DXA analysis for evidence of changes in unadjusted or body weight-adjusted BMD or BMC. Phenodeviant lines were identified based on statistical criteria that included a false discovery rate (FDR) <20% and Z-score >2.8. Genome-wide mapping scans were initiated on 22 lines, with evidence of high or low BMD or BMC that deviated by approximately -30% to +50% from the means. Several lines were discontinued as showing lack of heritability, but two heritable lines were identified with narrow chromosomal regions that allowed sequencing of potential mutant candidate genes. Novel mutations were identified in the Enpp1 (C397S) gene on chromosome 10 (line 4482) and the Ptpn6 (I482F) gene on chromosome 6 (line 4489) that were both associated with low bone mass. In addition, the phenotype of the Enpp1 mice showed a striking joint disease and calcification of blood vessels including the aorta, myocardium, and renal arteries and capillaries. These results support a role for the Enpp1 gene in the pathogenesis associated with mineralization of articular cartilage and vascular calcification. This work confirms the utility of the chemical mutagenesis approach for identification of potential disease genes and confirms the role of Enpp1 and Ptpn6 in regulating mineralization and skeletal bone mass.


Subject(s)
Bone Density/genetics , Calcinosis/genetics , Joint Diseases/genetics , Phosphoric Diester Hydrolases/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Pyrophosphatases/genetics , Vascular Diseases/genetics , Absorptiometry, Photon , Animals , Base Sequence , Chromosome Mapping , DNA Primers , Ethylnitrosourea/toxicity , Female , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mutagenesis , Mutagens/toxicity , Polymerase Chain Reaction
13.
J Bone Miner Res ; 24(7): 1234-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19257823

ABSTRACT

RANKL is an essential mediator of bone resorption, and its activity is inhibited by osteoprotegerin (OPG). Transgenic (Tg) rats were engineered to continuously overexpress OPG to study the effects of continuous long-term RANKL inhibition on bone volume, density, and strength. Lumbar vertebrae, femurs, and blood were obtained from 1-yr-old female OPG-Tg rats (n = 32) and from age-matched wildtype (WT) controls (n = 23). OPG-Tg rats had significantly greater serum OPG (up to 260-fold) and significantly lower serum TRACP5b and osteocalcin compared with WT controls. Vertebral histomorphometry showed significant reductions in osteoclasts and bone turnover parameters in OPG-Tg rats versus WT controls, and these reductions were associated with significantly greater peak load in vertebrae tested through compression. No apparent differences in bone material properties were observed in OPG-Tg rat vertebrae, based on their unchanged intrinsic strength parameters and their normal linear relationship between vertebral bone mass and strength. Femurs from OPG-Tg rats were of normal length but showed mild osteopetrotic changes, including reduced periosteal perimeter (-6%) and an associated reduction in bending strength. Serum OPG levels in WT rats showed no correlations with any measured parameter of bone turnover, mass, or strength, whereas the supraphysiological serum OPG levels in OPG-Tg rats correlated negatively with bone turnover parameters and positively with vertebral bone mass and strength parameters. In summary, low bone turnover after 1 yr of OPG overexpression in rats was associated with increased vertebral bone mass and proportional increases in bone strength, with no evidence for deleterious effects on vertebral material properties.


Subject(s)
Bone Density , Gene Expression , Lumbar Vertebrae/growth & development , Osteoprotegerin/biosynthesis , Animals , Bone Remodeling , Female , Lumbar Vertebrae/metabolism , Organ Size , Osteoclasts/metabolism , Osteopetrosis/metabolism , Osteoprotegerin/genetics , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Time Factors
14.
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
15.
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
16.
J Bone Miner Res ; 22(10): 1534-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17576164

ABSTRACT

UNLABELLED: Skeletal anabolism with PTH is achieved through daily injections that result in brief exposure to the peptide. We hypothesized that similar anabolic effects could be achieved with less frequent but more sustained exposures to PTH. A PTH-Fc fusion protein with a longer half-life than PTH(1-34) increased cortical and cancellous BMD and bone strength with once- or twice-weekly injections. INTRODUCTION: The anabolic effects of PTH are currently achieved with, and thought to require, daily injections that result in brief exposure to the peptide. We hypothesized that less frequent but more sustained exposures to PTH could also be anabolic for bone, provided that serum levels of PTH were not constant. MATERIALS AND METHODS: PTH(1-34) was fused to the Fc fragment of human IgG1 to increase the half-life of PTH. Skeletal anabolism was examined in mice and rats treated once or twice per week with this PTH-Fc fusion protein. RESULTS: PTH-Fc and PTH(1-34) had similar effects on PTH/PTHrP receptor activation, internalization, and signaling in vitro. However, PTH-Fc had a 33-fold longer mean residence time in the circulation of rats compared with that of PTH(1-34). Subcutaneous injection of PTH-Fc once or twice per week resulted in significant increases in bone volume, density, and strength in osteopenic ovariectomized mice and rats. These anabolic effects occurred in association with hypercalcemia and were significantly greater than those achievable with high concentrations of daily PTH(1-34). PTH-Fc also significantly improved cortical bone volume and density under conditions where daily PTH(1-34) did not. Antiresorptive co-therapy with estrogen further enhanced the ability of PTH-Fc to increase bone mass and strength in ovariectomized rats. CONCLUSIONS: These results challenge the notion that brief daily exposure to PTH is essential for its anabolic effects on cortical and cancellous bone. PTH-derived molecules with a sustained circulating half-life may represent a powerful and previously undefined anabolic regimen for cortical and cancellous bone.


Subject(s)
Anabolic Agents/administration & dosage , Anabolic Agents/pharmacology , Bone and Bones/drug effects , Immunoglobulin Fc Fragments/administration & dosage , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/pharmacology , Recombinant Proteins/pharmacology , Aging/physiology , Anabolic Agents/pharmacokinetics , Animals , Arrestins/metabolism , Bone and Bones/metabolism , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Estrogens/pharmacology , Half-Life , Humans , Male , Mice , Ovariectomy , Parathyroid Hormone/pharmacokinetics , Protein Transport , Rats , Receptor, Parathyroid Hormone, Type 1/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Time Factors , beta-Arrestins
17.
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
18.
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
19.
Crit Rev Eukaryot Gene Expr ; 13(2-4): 89-106, 2003.
Article in English | MEDLINE | ID: mdl-14696958

ABSTRACT

This study examined whether the calcium-sensing receptor (CaR) is expressed in normal adult human osteoblastic and osteoclastic cells in culture, and whether the calcimimetic, cinacalcet HCl (AMG 073), potentiates the effects of calcium (via CaR, or some other receptor/mechanism). When mouse or human osteoblastic cells were treated with higher concentrations of calcium (6.6 or 8.6 mM in alpha-MEM/10% FBS) than present in control cultures (1.6 mM), the previously well-documented increase in cell number was demonstrated. Cinacalcet HCl affected cell proliferation of CHO cells transfected with CaR, dose dependently, but had no effect on human or mouse osteoblastic cell proliferation in calcium-containing medium (1.6 or 8.6 mM). To test cinacalcet HCl and calcium on osteoclastic cells, peripheral blood mononuclear cells were cultured in medium containing RANK ligand and M-CSF, supplemented with calcium, and/or cinacalcet HCl. Tartrate-resistant acid phosphatase-positive multinucleated osteoclastic cells on plastic or bone were then counted at 11 and 21 days, respectively. Calcium (greater than 6.0 mM) inhibited osteoclast formation, but cinacalcet HCl (30-1000 nM) had no effect on osteoclastic formation or resorption in the presence of calcium (1.6 or 6.1 mM). RT-PCR did not detect CaR in human, rat, or mouse primary osteoblastic cells and cell lines or osteoclastic cells. In conclusion, these studies indicate that the calcium-induced increase in osteoblastic cell number, and the decrease in formation/function of osteoclastic cells, involves a mechanism or receptor other than CaR. In addition, the calcimimetic agent did not potentiate the effects of calcium on normal adult human bone cells in vitro.


Subject(s)
Calcium/metabolism , Naphthalenes/pharmacology , Osteoblasts/drug effects , Osteoclasts/drug effects , Receptors, Calcium-Sensing/biosynthesis , Animals , Bone and Bones/drug effects , CHO Cells , Cell Division , Cell Line , Cells, Cultured , Cinacalcet , Cricetinae , Culture Media/pharmacology , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Leukocytes, Mononuclear/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Mice , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...