Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bone ; 81: 562-572, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26318907

RESUMEN

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.


Asunto(s)
Resorción Ósea/prevención & control , Fragmentos Fc de Inmunoglobulinas/farmacología , Osteoprotegerina/farmacología , Proteínas Recombinantes de Fusión/farmacología , Vuelo Espacial , Ingravidez/efectos adversos , Fosfatasa Alcalina/sangre , Animales , Biomarcadores/sangre , Fenómenos Biomecánicos , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/etiología , Resorción Ósea/fisiopatología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Osteocalcina/sangre , Ligando RANK/antagonistas & inhibidores
2.
Bioorg Med Chem Lett ; 23(24): 6625-8, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24215889

RESUMEN

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.


Asunto(s)
Hiperparatiroidismo Secundario/tratamiento farmacológico , Tiazoles/química , Tiazoles/uso terapéutico , Urea/análogos & derivados , Administración Oral , Animales , Disponibilidad Biológica , Semivida , Hiperparatiroidismo Secundario/metabolismo , Hiperparatiroidismo Secundario/patología , Masculino , Hormona Paratiroidea/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores Sensibles al Calcio/química , Receptores Sensibles al Calcio/metabolismo , Tiazoles/farmacocinética
3.
J Pharmacol Exp Ther ; 337(3): 681-91, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21422163

RESUMEN

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.


Asunto(s)
Compuestos de Anilina/farmacología , Compuestos de Bifenilo/farmacología , Calcitonina/metabolismo , Calcio/agonistas , Calcio/metabolismo , Dietilaminas/farmacología , Hiperparatiroidismo Secundario/tratamiento farmacológico , Hormona Paratiroidea/metabolismo , Receptores Sensibles al Calcio/metabolismo , Animales , Compuestos de Bifenilo/administración & dosificación , Células CHO , Calcitonina/sangre , Calcio/sangre , Cricetinae , Cricetulus , Dietilaminas/administración & dosificación , Células HEK293 , Humanos , Hiperparatiroidismo Secundario/etiología , Hipocalcemia/complicaciones , Fosfatos de Inositol/metabolismo , Fallo Renal Crónico/complicaciones , Masculino , Glándulas Paratiroides/efectos de los fármacos , Hormona Paratiroidea/sangre , Fenetilaminas , Fosforilación/efectos de los fármacos , Propilaminas , Ratas , Ratas Sprague-Dawley , Diálisis Renal/efectos adversos
5.
J Med Chem ; 52(21): 6535-8, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19835382

RESUMEN

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.


Asunto(s)
Compuestos de Anilina/síntesis química , Metilaminas/síntesis química , Pirazoles/síntesis química , Receptores Sensibles al Calcio/agonistas , Administración Oral , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Animales , Disponibilidad Biológica , Línea Celular , Cristalografía por Rayos X , Humanos , Hiperparatiroidismo Secundario/tratamiento farmacológico , Masculino , Metilaminas/química , Metilaminas/farmacología , Estructura Molecular , Hormona Paratiroidea/sangre , Fenetilaminas , Propilaminas , Pirazoles/química , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Sensibles al Calcio/metabolismo , Estereoisomerismo , Relación Estructura-Actividad
6.
J Bone Miner Res ; 24(9): 1552-64, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19419305

RESUMEN

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.


Asunto(s)
Densidad Ósea/genética , Calcinosis/genética , Artropatías/genética , Hidrolasas Diéster Fosfóricas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Pirofosfatasas/genética , Enfermedades Vasculares/genética , Absorciometría de Fotón , Animales , Secuencia de Bases , Mapeo Cromosómico , Cartilla de ADN , Etilnitrosourea/toxicidad , Femenino , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mutagénesis , Mutágenos/toxicidad , Reacción en Cadena de la Polimerasa
7.
J Bone Miner Res ; 24(2): 182-95, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19016581

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Densidad Ósea/efectos de los fármacos , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/fisiopatología , Técnicas de Sustitución del Gen , Ligando RANK/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Afinidad de Anticuerpos/efectos de los fármacos , Especificidad de Anticuerpos/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/patología , Denosumab , Humanos , Hipercalcemia/tratamiento farmacológico , Ratones , Datos de Secuencia Molecular , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoprotegerina/metabolismo , Fenotipo , Unión Proteica/efectos de los fármacos , Ligando RANK/química , Ligando RANK/genética , Ligando RANK/farmacocinética , Ligando RANK/farmacología , Ligando RANK/uso terapéutico , Microtomografía por Rayos X
8.
J Bone Miner Res ; 24(4): 578-88, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19049336

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Proteínas Morfogenéticas Óseas/inmunología , Huesos/efectos de los fármacos , Marcadores Genéticos/inmunología , Osteogénesis/efectos de los fármacos , Osteoporosis Posmenopáusica/tratamiento farmacológico , Animales , Bioensayo , Fenómenos Biomecánicos , Densidad Ósea/efectos de los fármacos , Huesos/patología , Linaje de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fémur/efectos de los fármacos , Fémur/patología , Humanos , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Ratones , Pruebas de Neutralización , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteocalcina/sangre , Osteoporosis Posmenopáusica/sangre , Osteoporosis Posmenopáusica/patología , Osteoporosis Posmenopáusica/fisiopatología , Ovariectomía , Ratas , Ratas Sprague-Dawley , Tibia/efectos de los fármacos , Tibia/patología , Tomografía Computarizada por Rayos X
9.
J Appl Physiol (1985) ; 106(2): 582-95, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074574

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Vuelo Espacial , Ingravidez , Adaptación Fisiológica/genética , Animales , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica/métodos , Suspensión Trasera , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/fisiopatología , Atrofia Muscular/fisiopatología , Miostatina/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/genética , Reacción en Cadena de la Polimerasa , Proteínas Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Serina-Treonina Quinasas TOR , Factores de Tiempo
10.
J Bone Miner Res ; 23(6): 860-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18269310

RESUMEN

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.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Huesos/metabolismo , Eliminación de Gen , Osteogénesis , Proteínas Adaptadoras Transductoras de Señales , Animales , Biomarcadores/sangre , Densidad Ósea , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Huesos/diagnóstico por imagen , Calcio/sangre , Femenino , Marcadores Genéticos/genética , Glicoproteínas , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Noqueados , Fenotipo , Fosfatos/sangre , Estrés Mecánico , Tomografía Computarizada por Rayos X
11.
Circulation ; 117(3): 411-20, 2008 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-18172035

RESUMEN

BACKGROUND: The role of osteoprotegerin in vascular disease is unclear. Recent observational studies show that serum osteoprotegerin levels are associated with the severity and progression of coronary artery disease, atherosclerosis, and vascular calcification in patients. However, genetic and treatment studies in mice suggest that osteoprotegerin may protect against vascular calcification. METHODS AND RESULTS: To test whether osteoprotegerin induces or prevents vascular disease, we treated atherogenic diet-fed ldlr(-/-) mice with recombinant osteoprotegerin (Fc-OPG) or vehicle for 5 months. Vehicle-treated mice developed significant, progressive atherosclerosis with increased plasma osteoprotegerin levels, consistent with observational studies, and approximately 15% of these atherosclerotic lesions developed calcified cartilage-like metaplasia. Treatment with Fc-OPG significantly reduced the calcified lesion area without affecting atherosclerotic lesion size or number, vascular cytokines, or plasma cholesterol levels. Treatment also significantly reduced tissue levels of aortic osteocalcin, a marker of mineralization. CONCLUSIONS: These data support a role for osteoprotegerin in the vasculature as an inhibitor of calcification and a marker, rather than a mediator, of atherosclerosis.


Asunto(s)
Aterosclerosis/patología , Calcinosis/tratamiento farmacológico , Osteoprotegerina/farmacología , Enfermedades Vasculares/patología , Animales , Aorta , Aterosclerosis/etiología , Endotelio Vascular/química , Ratones , Ratones Noqueados , Osteoprotegerina/sangre , Ligando RANK/sangre , ARN Mensajero/sangre , Receptores de LDL/deficiencia , Proteínas Recombinantes , Enfermedades Vasculares/etiología
12.
Clin Exp Metastasis ; 25(2): 119-29, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18064531

RESUMEN

Bone metastases cause severe skeletal morbidity including fractures and hypercalcemia. Tumor cells in bone induce activation of osteoclasts, which mediate bone resorption and release of growth factors from bone matrix, resulting in a "vicious cycle" of bone breakdown and tumor proliferation. Receptor activator of NF-kappaB ligand (RANKL) is an essential mediator of osteoclast formation, function, and survival, and is blocked by a soluble decoy receptor, osteoprotegerin (OPG). In human malignancies that metastasize to bone, dysregulation of the RANK/RANKL/OPG pathway can increase the RANKL:OPG ratio, a condition which favors excessive osteolysis. In a mouse model of bone metastasis, RANKL protein levels in MDA-MB-231 (MDA-231) tumor-bearing bones were significantly higher than tumor-free bones. The resulting tumor-induced osteoclastogenesis and osteolysis was dose-dependently inhibited by recombinant OPG-Fc treatment, supporting the essential role for RANKL in this process. Using bioluminescence imaging in a mouse model of metastasis, we monitored the anti-tumor efficacy of RANKL inhibition on MDA-231 human breast cancer cells in a temporal manner. Treatment with OPG-Fc in vivo inhibited growth of MDA-231 tumor cells in bony sites when given both as a preventative (dosed day 0) and as a therapeutic agent for established bone metastases (dosed day 7). One mechanism by which RANKL inhibition reduced tumor burden appears to be indirect through inhibition of the "vicious cycle" and involved an increase in tumor cell apoptosis, as measured by active caspase-3. Here, we demonstrate for the first time that OPG-Fc treatment of mice with established bone metastases resulted in an overall improvement in survival.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias de la Mama/patología , Osteoprotegerina/farmacología , Ligando RANK/metabolismo , Animales , Apoptosis , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/mortalidad , Neoplasias Óseas/secundario , Huesos/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Osteoprotegerina/genética , Ligando RANK/antagonistas & inhibidores , Ligando RANK/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Trasplante Heterólogo
13.
J Bone Miner Res ; 22(10): 1534-47, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17576164

RESUMEN

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.


Asunto(s)
Anabolizantes/administración & dosificación , Anabolizantes/farmacología , Huesos/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/farmacología , Proteínas Recombinantes/farmacología , Envejecimiento/fisiología , Anabolizantes/farmacocinética , Animales , Arrestinas/metabolismo , Huesos/metabolismo , Línea Celular , Cricetinae , Relación Dosis-Respuesta a Droga , Estrógenos/farmacología , Semivida , Humanos , Masculino , Ratones , Ovariectomía , Hormona Paratiroidea/farmacocinética , Transporte de Proteínas , Ratas , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Factores de Tiempo , beta-Arrestinas
14.
J Biol Chem ; 282(29): 21237-43, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17522049

RESUMEN

Cardiovascular disease, such as atherosclerosis, has been associated with reduced bone mineral density and fracture risk. A major etiologic factor in atherogenesis is believed to be oxidized phospholipids. We previously found that these phospholipids inhibit spontaneous osteogenic differentiation of marrow stromal cells, suggesting that they may account for the clinical link between atherosclerosis and osteoporosis. Currently, anabolic agents that promote bone formation are increasingly used as a new treatment for osteoporosis. It is not known, however, whether atherogenic phospholipids alter the effects of bone anabolic agents, such as bone morphogenetic protein (BMP)-2 and parathyroid hormone (PTH). Therefore we investigated the effects of oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (ox-PAPC) on osteogenic signaling induced by BMP-2 and PTH in MC3T3-E1 cells. Results showed that ox-PAPC attenuated BMP-2 induction of osteogenic markers alkaline phosphatase and osteocalcin. Ox-PAPC also inhibited both spontaneous and BMP-induced expression of PTH receptor. Consistently, pretreatment of cells with ox-PAPC inhibited PTH-induced cAMP production and expression of immediate early genes Nurr1 and IL-6. Results from immunofluorescence and Western blot analyses showed that inhibitory effects of ox-PAPC on BMP-2 signaling were associated with inhibition of SMAD 1/5/8 but not p38-MAPK activation. These effects appear to be due to ox-PAPC activation of the ERK pathway, as the ERK inhibitor PD98059 reversed ox-PAPC inhibitory effects on BMP-2-induced alkaline phosphatase activity, osteocalcin expression, and SMAD activation. These results suggest that atherogenic lipids inhibit osteogenic signaling induced by BMP-2 and PTH, raising the possibility that hyperlipidemia and atherogenic phospholipids may interfere with anabolic therapy.


Asunto(s)
Aterosclerosis/patología , Proteínas Morfogenéticas Óseas/metabolismo , Osteoblastos/metabolismo , Hormona Paratiroidea/metabolismo , Fosfolípidos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Aterosclerosis/metabolismo , Proteína Morfogenética Ósea 2 , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Ratones , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Fosfatidilcolinas/metabolismo , Fosforilcolina/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Bone ; 39(4): 754-66, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16730481

RESUMEN

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.


Asunto(s)
Enfermedades Óseas Metabólicas/fisiopatología , Huesos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas Wnt/fisiología , Células 3T3 , Animales , Densidad Ósea , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Huesos/patología , Huesos/fisiopatología , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas Relacionadas con Receptor de LDL/metabolismo , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/sangre , Osteogénesis/genética , Osteogénesis/fisiología , Embarazo , Ratas , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo
16.
Nature ; 440(7084): 692-6, 2006 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-16572175

RESUMEN

Bone metastases are a frequent complication of many cancers that result in severe disease burden and pain. Since the late nineteenth century, it has been thought that the microenvironment of the local host tissue actively participates in the propensity of certain cancers to metastasize to specific organs, and that bone provides an especially fertile 'soil'. In the case of breast cancers, the local chemokine milieu is now emerging as an explanation for why these tumours preferentially metastasize to certain organs. However, as the inhibition of chemokine receptors in vivo only partially blocks metastatic behaviour, other factors must exist that regulate the preferential metastasis of breast cancer cells. Here we show that the cytokine RANKL (receptor activator of NF-kappaB ligand) triggers migration of human epithelial cancer cells and melanoma cells that express the receptor RANK. RANK is expressed on cancer cell lines and breast cancer cells in patients. In a mouse model of melanoma metastasis, in vivo neutralization of RANKL by osteoprotegerin results in complete protection from paralysis and a marked reduction in tumour burden in bones but not in other organs. Our data show that local differentiation factors such as RANKL have an important role in cell migration and the tissue-specific metastatic behaviour of cancer cells.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Proteínas Portadoras/metabolismo , Movimiento Celular , Glicoproteínas de Membrana/metabolismo , Metástasis de la Neoplasia/patología , Animales , Neoplasias Óseas/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Portadoras/genética , Muerte Celular , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Células Epiteliales/patología , Femenino , Humanos , Melanoma/metabolismo , Melanoma/patología , Glicoproteínas de Membrana/genética , Ratones , Especificidad de Órganos , Parálisis , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Transducción de Señal
17.
Endocrinology ; 146(8): 3235-43, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15845617

RESUMEN

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.


Asunto(s)
Adenocarcinoma/prevención & control , Resorción Ósea/prevención & control , Proteínas Portadoras/antagonistas & inhibidores , Neoplasias del Colon/prevención & control , Difosfonatos/farmacología , Glicoproteínas/farmacología , Hipercalcemia/prevención & control , Glicoproteínas de Membrana/antagonistas & inhibidores , Adenocarcinoma/sangre , Animales , Antineoplásicos/farmacología , Calcio/sangre , Línea Celular Tumoral , Neoplasias del Colon/sangre , Modelos Animales de Enfermedad , Humanos , Hipercalcemia/etiología , Ligandos , Ratones , FN-kappa B/metabolismo , Osteoprotegerina , Pamidronato , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares , Receptores del Factor de Necrosis Tumoral
18.
Bone ; 34(4): 656-64, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15050896

RESUMEN

Osteoporosis is a chronic condition that is typically treated by the long-term repeated administration of antiresorptive agents. Gene therapy has the potential to deliver protein-based antiresorptive agents without the need for repeated administration. Osteoprotegerin (OPG) is a naturally occuring protein that prevents bone resorption by inhibiting osteoclast formation, function and survival. We tested whether adeno-associated virus (AAV) could deliver OPG at levels that are sufficient to reverse established osteopenia in ovariectomized (OVX) mice without causing liver toxicity. Tibial bone mineral density (BMD) was measured by peripheral quantitative computed tomography (pQCT) in 12-week-old CDF1 mice prior to OVX or sham surgery. Six weeks later, BMD was significantly reduced in OVX mice compared to sham controls or pre-surgery values. Sham and OVX mice were then injected once IV with an AAV vector carrying cDNA for recombinant hOPG (AAV-OPG) or beta-galactosidase (AAV-betaGal). BMD and bone histomorphometry were assessed 10 weeks after treatment. A single injection of AAV-OPG led to the appearance of human OPG (hOPG) in the serum of mice within 7 days, and high serum levels of hOPG were maintained for the duration of the 10-week study. At the end of the study, OVX mice given AAV-OPG had significantly greater tibial BMD compared to age-matched OVX animals given AAV-betaGal. In sham-operated mice, AAV-OPG also significantly increased tibial BMD compared to AAV-betaGal. The increased BMD in AAV-OPG animals was accompanied by significantly increased bone volume and significantly reduced osteoclast surfaces in the proximal tibial metaphysis. Liver histology was normal, and circulating activities of hepatocyte cytosolic enzymes were unaffected by AAV exposure. In an accompanying experiment, young (3-4 weeks) C57BL/6 mice treated once IV with AAV-OPG maintained pharmacologically active levels of OPG in serum for at least 16 months. In summary, a single AAV-OPG treatment reversed established osteopenia in OVX mice without evidence of liver toxicity. AAV delivery appears to be a safe and effective method for producing sustained systemic exposure to OPG.


Asunto(s)
Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/terapia , Terapia Genética , Glicoproteínas/genética , Glicoproteínas/uso terapéutico , Ovariectomía , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Animales , Densidad Ósea/fisiología , Enfermedades Óseas Metabólicas/patología , Resorción Ósea/genética , Resorción Ósea/prevención & control , Resorción Ósea/terapia , Dependovirus/genética , Dependovirus/fisiología , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/genética , Glicoproteínas/efectos adversos , Glicoproteínas/metabolismo , Humanos , Hígado/efectos de los fármacos , Ratones , Osteoprotegerina , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/sangre , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Tibia/fisiopatología , Factores de Tiempo
19.
Arterioscler Thromb Vasc Biol ; 24(2): e6-10, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14670933

RESUMEN

OBJECTIVE: Osteoporosis is associated epidemiologically with atherosclerosis and hyperlipidemia. We previously found that atherogenic lipids regulate bone formation. To determine whether hyperlipidemia also affects bone resorption, we compared osteoclastogenesis in marrow preosteoclasts derived from hyperlipidemic versus control mice. METHODS: Nonadherent marrow cells from low-density lipoprotein receptor-/- (LDLR-/-)and C57BL/6J mice were cultured with M-CSF and ligand for receptor activator of nuclear factor-kappaB (RANKL). Functional osteoclastic activity, measured as number of resorption pits, was significantly greater in 12-month-old LDLR-/-. Similar results were obtained in 5- and 10-month-old LDLR-/- versus C57BL/6J mice on a high-fat diet. Osteoclastic differentiation, indicated by tartrate resistant acid phosphatase (TRAP) activity, was significantly greater in the 12-month-old LDLR-/-, and there was a trend toward increased TRAP activity in LDLR-/- on a high-fat diet, at ages 5 and 10 months. Osteoclastic parameters correlated with total serum lipoproteins with a possible threshold effect. Osteoporotic human cortical bone stained positive for lipids in the perivascular space of Haversian canals by oil red O. The presence of lipid hydroperoxides was detected in bone marrow from hyperlipidemic mice. CONCLUSIONS: Hyperlipidemia may contribute to osteoporosis via increased osteoclastic bone resorption.


Asunto(s)
Células de la Médula Ósea/patología , Diferenciación Celular/fisiología , Hiperlipidemias/patología , Osteoclastos/patología , Animales , Resorción Ósea/patología , Células Cultivadas , Dieta Aterogénica , Hiperlipidemias/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Osteoporosis/etiología , Osteoporosis/patología , Receptores de LDL/deficiencia , Células Madre/patología
20.
J Bone Miner Res ; 18(5): 852-8, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12733724

RESUMEN

Osteoprotegerin (OPG) is a naturally occurring negative regulator of osteoclast differentiation, activation, and survival. We created a recombinant form of human OPG (rhOPG), with a sustained serum half-life, to achieve prolonged antiresorptive activity. This study describes the rapid and sustained antiresorptive effects that are achieved with a single treatment with rhOPG. Male Sprague-Dawley rats (10 weeks old) were given a single bolus intravenous injection of vehicle (PBS) or rhOPG (5 mg/kg). PBS- and rhOPG-treated rats (n = 6/group) were killed at 0, 0.5, 1, 2, 5, 10, 20, and 30 days post-treatment. rhOPG-treated rats were compared with their age-matched controls. The main pharmacologic effect of rhOPG was a rapid (24 h) reduction in osteoclast surface in the tibia, which reached a nadir on days 5 and 10 (95% reduction vs. vehicle controls). Osteoclast surface remained significantly reduced 30 days after the single treatment with rhOPG. Tibial cancellous bone volume was significantly increased within 5 days of rhOPG treatment (23%) and reached a peak increase of 58% on day 30. Femoral bone mineral density was significantly increased in rhOPG-treated rats on days 10 and 20. Pharmacokinetic analysis revealed that serum concentrations of rhOPG remained at measurable levels throughout the 30-day study. These data show that a single intravenous injection of rhOPG in young growing rats causes significant gains in bone volume and density, which are associated with rapid and sustained suppression of osteoclastic bone resorption.


Asunto(s)
Resorción Ósea , Glicoproteínas/farmacología , Animales , Femenino , Glicoproteínas/sangre , Glicoproteínas/farmacocinética , Humanos , Masculino , Osteoprotegerina , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/sangre , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...