Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Bone ; 81: 562-572, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26318907

ABSTRACT

Bone loss associated with microgravity exposure poses a significant barrier to long-duration spaceflight. Osteoprotegerin-Fc (OPG-Fc) is a receptor activator of nuclear factor kappa-B ligand (RANKL) inhibitor that causes sustained inhibition of bone resorption after a single subcutaneous injection. We tested the ability of OPG-Fc to preserve bone mass during 12 days of spaceflight (SF). 64-day-old female C57BL/6J mice (n=12/group) were injected subcutaneously with OPG-Fc (20mg/kg) or an inert vehicle (VEH), 24h prior to launch. Ground control (GC) mice (VEH or OPG-Fc) were maintained under environmental conditions that mimicked those in the space shuttle middeck. Age-matched baseline (BL) controls were sacrificed at launch. GC/VEH, but not SF/VEH mice, gained tibia BMD and trabecular volume fraction (BV/TV) during the mission (P<0.05 vs. BL). SF/VEH mice had lower BV/TV vs. GC/VEH mice, while SF/OPG-Fc mice had greater BV/TV than SF/VEH or GC/VEH. SF reduced femur elastic and maximum strength in VEH mice, with OPG-Fc increasing elastic strength in SF mice. Serum TRAP5b was elevated in SF/VEH mice vs. GC/VEH mice. Conversely, SF/OPG-Fc mice had lower TRAP5b levels, suggesting that OPG-Fc preserved bone during spaceflight via inhibition of osteoclast-mediated bone resorption. Decreased bone formation also contributed to the observed osteopenia, based on the reduced femur periosteal bone formation rate and serum osteocalcin level. Overall, these observations suggest that the beneficial effects of OPG-Fc during SF are primarily due to dramatic and sustained suppression of bone resorption. In growing mice, this effect appears to compensate for the SF-related inhibition of bone formation, while preventing any SF-related increase in bone resorption. We have demonstrated that the young mouse is an appropriate new model for SF-induced osteopenia, and that a single pre-flight treatment with OPG-Fc can effectively prevent the deleterious effects of SF on mouse bone.


Subject(s)
Bone Resorption/prevention & control , Immunoglobulin Fc Fragments/pharmacology , Osteoprotegerin/pharmacology , Recombinant Fusion Proteins/pharmacology , Space Flight , Weightlessness/adverse effects , Alkaline Phosphatase/blood , Animals , Biomarkers/blood , Biomechanical Phenomena , Bone Density/drug effects , Bone Density Conservation Agents/pharmacology , Bone Resorption/etiology , Bone Resorption/physiopathology , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Osteocalcin/blood , RANK Ligand/antagonists & inhibitors
2.
Bioorg Med Chem Lett ; 23(24): 6625-8, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24215889

ABSTRACT

A series of urea based calcimimetics was optimized for potency and oral bioavailability. Crucial to this process was overcoming the poor pharmacokinetic properties of lead thiazole 1. Metabolism-guided modifications, characterized by the use of metabolite identification (ID) and measurement of time dependent inhibition (TDI) of CYP3A4, were essential to finding a compound suitable for oral dosing. Calcimimetic 18 exhibited excellent in vivo potency in a 5/6 nephrectomized rat model and cross-species pharmacokinetics.


Subject(s)
Hyperparathyroidism, Secondary/drug therapy , Thiazoles/chemistry , Thiazoles/therapeutic use , Urea/analogs & derivatives , Administration, Oral , Animals , Biological Availability , Half-Life , Hyperparathyroidism, Secondary/metabolism , Hyperparathyroidism, Secondary/pathology , Male , Parathyroid Hormone/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Thiazoles/pharmacokinetics
3.
J Pharmacol Exp Ther ; 337(3): 681-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21422163

ABSTRACT

Calcimimetics are positive allosteric modulators to the calcium-sensing receptor (CaSR). Activation of the CaSR inhibits the secretion of parathyroid hormone (PTH), stimulates the secretion of calcitonin, and decreases serum calcium (Ca(2+)). Cinacalcet, a second-generation calcimimetic, is used therapeutically to control PTH in patients with chronic kidney disease who are on dialysis with secondary hyperparathyroidism. A calcimimetic that displays increased separation of PTH versus Ca(2+) lowering in patients would potentially allow the use of calcimimetics to treat patients in earlier stages of renal disease because hypocalcemia can develop in this population. Toward this end, we developed a third-generation calcimimetic, determined the molecular pharmacological properties of it using an operation model of allosteric modulation/agonism, and measured the compound effects on PTH, serum ionized Ca(2+), and calcitonin levels in 5/6 nephrectomized rats. We found the new molecule effectively reduced PTH levels without promoting calcitonin secretion or hypocalcemia. Furthermore, our third-generation molecule was less efficacious at promoting calcitonin secretion from human thyroid carcinoma cells compared with 3-(2-chlorophenyl)-N-((1R)-1-(3-methoxyphenyl)ethyl)-1-propanamine (R-568), a first-generation calcimimetic. These data provide evidence that calcimimetics with increased potency can be used to lower PTH without production of significant hypocalcemia because the threshold for inhibition of PTH secretion is much lower than the threshold for calcitonin secretion.


Subject(s)
Aniline Compounds/pharmacology , Biphenyl Compounds/pharmacology , Calcitonin/metabolism , Calcium/agonists , Calcium/metabolism , Diethylamines/pharmacology , Hyperparathyroidism, Secondary/drug therapy , Parathyroid Hormone/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Biphenyl Compounds/administration & dosage , CHO Cells , Calcitonin/blood , Calcium/blood , Cricetinae , Cricetulus , Diethylamines/administration & dosage , HEK293 Cells , Humans , Hyperparathyroidism, Secondary/etiology , Hypocalcemia/complications , Inositol Phosphates/metabolism , Kidney Failure, Chronic/complications , Male , Parathyroid Glands/drug effects , Parathyroid Hormone/blood , Phenethylamines , Phosphorylation/drug effects , Propylamines , Rats , Rats, Sprague-Dawley , Renal Dialysis/adverse effects
5.
J Med Chem ; 52(21): 6535-8, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19835382

ABSTRACT

Our efforts to discover potent, orally bioavailable type II calcimimetic agents for the treatment of secondary hyperparathyroidism focused on the development of ring constrained analogues of the known calcimimetic R-568. The structure-activity relationships of various substituted heterocycles and their effects on the human calcium-sensing receptor are discussed. Pyrazole 15 was shown to be efficacious in a rat in vivo pharmacodynamic model.


Subject(s)
Aniline Compounds/chemical synthesis , Methylamines/chemical synthesis , Pyrazoles/chemical synthesis , Receptors, Calcium-Sensing/agonists , Administration, Oral , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Biological Availability , Cell Line , Crystallography, X-Ray , Humans , Hyperparathyroidism, Secondary/drug therapy , Male , Methylamines/chemistry , Methylamines/pharmacology , Molecular Structure , Parathyroid Hormone/blood , Phenethylamines , Propylamines , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/metabolism , Stereoisomerism , Structure-Activity Relationship
6.
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
7.
J Appl Physiol (1985) ; 106(2): 582-95, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074574

ABSTRACT

Spaceflight results in a number of adaptations to skeletal muscle, including atrophy and shifts toward faster muscle fiber types. To identify changes in gene expression that may underlie these adaptations, we used both microarray expression analysis and real-time polymerase chain reaction to quantify shifts in mRNA levels in the gastrocnemius from mice flown on the 11-day, 19-h STS-108 shuttle flight and from normal gravity controls. Spaceflight data also were compared with the ground-based unloading model of hindlimb suspension, with one group of pure suspension and one of suspension followed by 3.5 h of reloading to mimic the time between landing and euthanization of the spaceflight mice. Analysis of microarray data revealed that 272 mRNAs were significantly altered by spaceflight, the majority of which displayed similar responses to hindlimb suspension, whereas reloading tended to counteract these responses. Several mRNAs altered by spaceflight were associated with muscle growth, including the phosphatidylinositol 3-kinase regulatory subunit p85alpha, insulin response substrate-1, the forkhead box O1 transcription factor, and MAFbx/atrogin1. Moreover, myostatin mRNA expression tended to increase, whereas mRNA levels of the myostatin inhibitor FSTL3 tended to decrease, in response to spaceflight. In addition, mRNA levels of the slow oxidative fiber-associated transcriptional coactivator peroxisome proliferator-associated receptor (PPAR)-gamma coactivator-1alpha and the transcription factor PPAR-alpha were significantly decreased in spaceflight gastrocnemius. Finally, spaceflight resulted in a significant decrease in levels of the microRNA miR-206. Together these data demonstrate that spaceflight induces significant changes in mRNA expression of genes associated with muscle growth and fiber type.


Subject(s)
Gene Expression Regulation , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Space Flight , Weightlessness , Adaptation, Physiological/genetics , Animals , Cluster Analysis , Female , Gene Expression Profiling/methods , Hindlimb Suspension , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Myostatin/genetics , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/metabolism , Reproducibility of Results , TOR Serine-Threonine Kinases , Time Factors
8.
J Bone Miner Res ; 24(4): 578-88, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19049336

ABSTRACT

The development of bone-rebuilding anabolic agents for potential use in the treatment of bone loss conditions, such as osteoporosis, has been a long-standing goal. Genetic studies in humans and mice have shown that the secreted protein sclerostin is a key negative regulator of bone formation, although the magnitude and extent of sclerostin's role in the control of bone formation in the aging skeleton is still unclear. To study this unexplored area of sclerostin biology and to assess the pharmacologic effects of sclerostin inhibition, we used a cell culture model of bone formation to identify a sclerostin neutralizing monoclonal antibody (Scl-AbII) for testing in an aged ovariectomized rat model of postmenopausal osteoporosis. Six-month-old female rats were ovariectomized and left untreated for 1 yr to allow for significant estrogen deficiency-induced bone loss, at which point Scl-AbII was administered for 5 wk. Scl-AbII treatment in these animals had robust anabolic effects, with marked increases in bone formation on trabecular, periosteal, endocortical, and intracortical surfaces. This not only resulted in complete reversal, at several skeletal sites, of the 1 yr of estrogen deficiency-induced bone loss, but also further increased bone mass and bone strength to levels greater than those found in non-ovariectomized control rats. Taken together, these preclinical results establish sclerostin's role as a pivotal negative regulator of bone formation in the aging skeleton and, furthermore, suggest that antibody-mediated inhibition of sclerostin represents a promising new therapeutic approach for the anabolic treatment of bone-related disorders, such as postmenopausal osteoporosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Bone Morphogenetic Proteins/immunology , Bone and Bones/drug effects , Genetic Markers/immunology , Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Animals , Biological Assay , Biomechanical Phenomena , Bone Density/drug effects , Bone and Bones/pathology , Cell Lineage/drug effects , Disease Models, Animal , Female , Femur/drug effects , Femur/pathology , Humans , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/pathology , Mice , Neutralization Tests , Organ Size/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/blood , Osteoporosis, Postmenopausal/blood , Osteoporosis, Postmenopausal/pathology , Osteoporosis, Postmenopausal/physiopathology , Ovariectomy , Rats , Rats, Sprague-Dawley , Tibia/drug effects , Tibia/pathology , Tomography, X-Ray Computed
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(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
SELECTION OF CITATIONS
SEARCH DETAIL