Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Numer Method Biomed Eng ; 30(1): 1-27, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24039120

RESUMEN

Denosumab, a fully human monoclonal antibody, has been approved for the treatment of postmenopausal osteoporosis. The therapeutic effect of denosumab rests on its ability to inhibit osteoclast differentiation. Here, we present a computational approach on the basis of coupling a pharmacokinetics model of denosumab with a pharmacodynamics model for quantifying the effect of denosumab on bone remodeling. The pharmacodynamics model comprises an integrated systems biology-continuum micromechanics approach, including a bone cell population model, considering the governing biochemical factors of bone remodeling (including the action of denosumab), and a multiscale micromechanics-based bone mechanics model, for implementing the mechanobiology of bone remodeling in our model. Numerical studies of postmenopausal osteoporosis show that denosumab suppresses osteoclast differentiation, thus strongly curtailing bone resorption. Simulation results also suggest that denosumab may trigger a short-term bone volume gain, which is, however, followed by constant or decreasing bone volume. This evolution is accompanied by a dramatic decrease of the bone turnover rate by more than one order of magnitude. The latter proposes dominant occurrence of secondary mineralization (which is not anymore impeded through cellular activity), leading to higher mineral concentration per bone volume. This explains the overall higher bone mineral density observed in denosumab-related clinical studies.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Simulación por Computador , Modelos Teóricos , Osteoporosis Posmenopáusica/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Densidad Ósea/efectos de los fármacos , Remodelación Ósea/efectos de los fármacos , Resorción Ósea/tratamiento farmacológico , Huesos/efectos de los fármacos , Huesos/metabolismo , Calibración , Denosumab , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Posmenopausia
2.
Acta Biomater ; 9(6): 7014-24, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23467040

RESUMEN

During the past two decades, research on ceramic scaffolds for bone regeneration has progressed rapidly; however, currently available porous scaffolds remain unsuitable for load-bearing applications. The key to success is to apply microstructural design strategies to develop ceramic scaffolds with mechanical properties approaching those of bone. Here we report on the development of a unique microstructurally designed ceramic scaffold, strontium-hardystonite-gahnite (Sr-HT-gahnite), with 85% porosity, 500µm pore size, a competitive compressive strength of 4.1±0.3MPa and a compressive modulus of 170±20MPa. The in vitro biocompatibility of the scaffolds was studied using primary human bone-derived cells. The ability of Sr-HT-gahnite scaffolds to repair critical-sized bone defects was also investigated in a rabbit radius under normal load, with ß-tricalcium phosphate/hydroxyapatite scaffolds used in the control group. Studies with primary human osteoblast cultures confirmed the bioactivity of these scaffolds, and regeneration of rabbit radial critical defects demonstrated that this material induces new bone defect bridging, with clear evidence of regeneration of original radial architecture and bone marrow environment.


Asunto(s)
Regeneración Ósea/fisiología , Sustitutos de Huesos/síntesis química , Cerámica/química , Regeneración Tisular Dirigida/instrumentación , Fracturas del Radio/fisiopatología , Fracturas del Radio/cirugía , Andamios del Tejido , Animales , Diseño de Equipo , Análisis de Falla de Equipo , Curación de Fractura/fisiología , Regeneración Tisular Dirigida/métodos , Masculino , Ensayo de Materiales , Conejos , Fracturas del Radio/diagnóstico , Resultado del Tratamiento
3.
Acta Biomater ; 8(11): 4162-72, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22842031

RESUMEN

This is the first reported study to prepare highly porous baghdadite (Ca3ZrSi2O9) scaffolds with and without surface modification and investigate their ability to repair critical-sized bone defects in a rabbit radius under normal load. The modification was carried out to improve the mechanical properties of the baghdadite scaffolds (particularly to address their brittleness) by coating their surfaces with a thin layer (∼400 nm) of polycaprolactone (PCL)/bioactive glass nanoparticles (nBGs). The ß-tricalcium phosphate/hydroxyapatite (TCP/HA) scaffolds with and without modification were used as the control groups. All of the tested scaffolds had an open and interconnected porous structure with a porosity of ∼85% and average pore size of 500 µm. The scaffolds (six per scaffold type and size of 4 mm × 4 mm × 15 mm) were implanted (press-fit) into the rabbit radial segmental defects for 12 weeks. Micro-computed tomography and histological evaluations were used to determine bone ingrowth, bone quality, and implant integration after 12 weeks of healing. Extensive new bone formation with complete bridging of the radial defect was evident with the baghdadite scaffolds (modified/unmodified) at the periphery and in close proximity to the ceramics within the pores, in contrast to TCP/HA scaffolds (modified/unmodified), where bone tended to grow between the ulna adjacent to the implant edge. Although the modification of the baghdadite scaffolds significantly improved their mechanical properties, it did not show any significant effect on in vivo bone formation. Our findings suggest that baghdadite scaffolds with and without modification can serve as a potential material to repair critical sized bone defects.


Asunto(s)
Huesos/patología , Silicatos/química , Andamios del Tejido/química , Cicatrización de Heridas , Animales , Líquidos Corporales , Regeneración Ósea/efectos de los fármacos , Huesos/diagnóstico por imagen , Huesos/efectos de los fármacos , Fosfatos de Calcio/farmacología , Modelos Animales de Enfermedad , Durapatita/farmacología , Masculino , Fenómenos Mecánicos/efectos de los fármacos , Peso Molecular , Osteoclastos/metabolismo , Osteoclastos/patología , Poliésteres/farmacología , Porosidad , Conejos , Soluciones , Cicatrización de Heridas/efectos de los fármacos , Microtomografía por Rayos X
4.
Int J Biochem Cell Biol ; 39(6): 1077-81, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17174136

RESUMEN

RANK ligand (RANKL), a key mediator of bone resorption in normal and pathological states, is expressed as membrane-bound or soluble forms by tissues as diverse as lymph nodes, spleen, thymus and bone-forming cells. In normal bone turnover and in bone metastasis, RANKL stimulates the formation and activity of bone-removing cells, osteoclasts, by binding to its cognate receptor, RANK, on osteoclasts and their progenitors; these processes are disrupted by binding of RANKL to osteoprotegerin (OPG), a soluble decoy receptor. Whilst no mutations in the RANKL gene have yet been identified in human disease, mutations that result in enhanced RANK signalling through inactivation of OPG or activation of RANK are associated with Juvenile Paget's disease and familial expansile osteolysis, respectively. This review focuses on the central role of RANKL in bone resorption and on the therapeutic targeting of RANKL in osteoporosis, humoral hypercalcaemia of malignancy and bone metastasis.


Asunto(s)
Ligando RANK/genética , Ligando RANK/metabolismo , Secuencia de Aminoácidos , Animales , Resorción Ósea/metabolismo , Resorción Ósea/prevención & control , Perfilación de la Expresión Génica , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Osteoprotegerina/metabolismo , Ligando RANK/antagonistas & inhibidores , Homología de Secuencia de Aminoácido
6.
Arthritis Rheum ; 54(6): 1772-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16736519

RESUMEN

OBJECTIVE: Rheumatoid arthritis (RA) is a chronic inflammatory disease that may result in debilitating joint deformities with destruction of bone and cartilage. Inflammation is still considered the pivotal inducer of both components of joint damage. Results of recent animal studies suggested a prominent contribution of osteoclastic bone resorption that could be dissociated from inflammation. RANKL and its natural decoy receptor, osteoprotegerin (OPG), play key roles in osteoclast activation. In a group of patients with early RA not treated with disease-modifying drugs, we tested the hypothesis that osteoclast activation, reflected by the serum OPG:RANKL ratio at baseline, is negatively associated with progression of bone damage, independent of inflammation. METHODS: OPG and RANKL levels, together with a parameter of inflammation (first-year time-averaged erythrocyte sedimentation rate [tESR]), were measured in 92 patients with newly diagnosed early active RA who were participants in a randomized study. The tESR and the OPG:RANKL ratio were evaluated for the ability to predict 5-year radiographic progression of joint damage. RESULTS: The first-year tESR and the OPG:RANKL ratio, as measured at baseline, independently predicted 5-year radiographic progression of joint damage (both P < or = 0.001). Progression of radiographic damage was greatest in patients with a high tESR and a low OPG:RANKL ratio and was lowest in patients with a low tESR and a high OPG:RANKL ratio. CONCLUSION: This study in patients with early untreated RA is the first to confirm the findings in animal models of arthritis, that radiographic progression of the bone component of joint destruction is dependent on both inflammation (tESR) and osteoclast activation (the OPG:RANKL ratio).


Asunto(s)
Artritis Reumatoide/sangre , Artritis Reumatoide/patología , Proteínas Portadoras/sangre , Glicoproteínas/sangre , Glicoproteínas de Membrana/sangre , Receptores Citoplasmáticos y Nucleares/sangre , Receptores del Factor de Necrosis Tumoral/sangre , Artritis Reumatoide/diagnóstico por imagen , Artrografía , Sedimentación Sanguínea , Progresión de la Enfermedad , Femenino , Humanos , Articulaciones/patología , Masculino , Persona de Mediana Edad , Osteoclastos/fisiología , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B
7.
Bone ; 37(1): 87-95, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15869920

RESUMEN

The combination of PTH with OPG has been proposed as a potential therapy in patients with severe osteoporosis. In the present study, we examined the bone material of aged ovariectomized (OVX) rats treated either with PTH (1-34) or OPG alone or in combination of both. The micro- and nanostructural characteristics of the mineralized bone were evaluated using quantitative backscattered electron imaging (qBEI) and small-angle X-ray scattering (SAXS). Rats (n=68) were either sham-operated or ovariectomized (OVX) at the age of 3 months, and 15 months later, OVX animals were treated either with vehicle, OPG (10 mg/kg), PTH (80 microg/kg) or a combination of both during 5.5 months. All treatments were by subcutaneous injection, 3 days per week. Secondary metaphyseal spongiosa from distal femora was assessed for mineralized bone volume (BV/TV), for the mean Ca-concentration (Camean), the width of the bone mineralization density distribution (Cawidth), as well as the average mineral particle thickness parameter (T) and the degree of alignment of the mineral particles (rho). A remarkable increase of BV/TV up to 139% (P<0.001) was observed in the PTH-treated groups independently of OPG. Camean was slightly increased (+1.7%, P<0.05) in the OPG-treated group. Cawidth was reduced (-6.4%, P<0.01, and -8.9%, P<0.001) in animals treated with OPG and PTH+OPG, respectively. In contrast, Cawidth in sham-operated rats was 16.0% (P<0.001) higher than in OVX. The T parameter was not altered in the trabecular bone within the group of treated and untreated OVX rats. However, the non-ovariectomized animals exhibited a significantly lower T value (-7.1%, P<0.01) with respect to OVX. In conclusion, qBEI and SAXS data of OVX rats suggest that PTH alone was responsible for increase of bone volume, whereas OPG positively influenced the homogeneity and density of mineralization without affecting the nanostructure of the bone material.


Asunto(s)
Huesos/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Glicoproteínas/farmacología , Ovariectomía , Teriparatido/farmacología , Animales , Huesos/química , Huesos/patología , Calcio/análisis , Quimioterapia Combinada , Microanálisis por Sonda Electrónica , Fémur/química , Fémur/efectos de los fármacos , Fémur/patología , Glicoproteínas/uso terapéutico , Humanos , Minerales/análisis , Minerales/química , Osteoporosis/tratamiento farmacológico , Osteoprotegerina , Ratas , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Teriparatido/uso terapéutico
8.
Clin Exp Metastasis ; 21(5): 381-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15672862

RESUMEN

Osteoprotegerin (OPG) plays a central role in controlling bone resorption. Exogenous administration of OPG has been shown to be effective in preventing osteolysis and limiting the growth of osteolytic metastasis. The objective of this study was to investigate the effects of OPG on osteoblastic prostate cancer (CaP) metastases in an animal model. LuCaP 23.1 cells were injected intra-tibially and Fc-OPG (6.0 mg/kg) was administered subcutaneously three times a week starting either 24 hours prior to cell injection (prevention regimen) or at 4 weeks post-injection (treatment regimen). Changes in bone mineral density at the tumor site were determined by dual x-ray absorptiometry. Tumor growth was monitored by evaluating serum prostate specific antigen (PSA). Fc-OPG did not inhibit establishment of osteoblastic bone lesions of LuCaP 23.1, but it decreased growth of the tumor cells, as determined by decreases in serum PSA levels of 73.0 +/- 44.3% (P < 0.001) and 78.3 +/- 25.3% (P < 0.001) under the treatment and prevention regimens, respectively, compared to the untreated tumor-bearing animals. Administration of Fc-OPG decreased the proliferative index by 35.0% (P = 0.1838) in the treatment group, and 75.2% (P = 0.0358) in the prevention group. The results of this study suggest a potential role for OPG in the treatment of established osteoblastic CaP bone metastases.


Asunto(s)
Neoplasias Óseas/terapia , Glicoproteínas/administración & dosificación , Osteoblastos/metabolismo , Neoplasias de la Próstata/terapia , Receptores Citoplasmáticos y Nucleares/administración & dosificación , Tibia/patología , Animales , Densidad Ósea , Neoplasias Óseas/secundario , Inyecciones Subcutáneas , Metástasis Linfática/patología , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Osteoprotegerina , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/patología , Receptores del Factor de Necrosis Tumoral/administración & dosificación , Tibia/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Blood ; 98(13): 3534-40, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11739154

RESUMEN

Multiple myeloma is a B-cell malignancy characterized by the accumulation of plasma cells in the bone marrow and the development of osteolytic bone disease. The present study demonstrates that myeloma cells express the critical osteoclastogenic factor RANKL (the ligand for receptor activator of NF-kappa B). Injection of 5T2MM myeloma cells into C57BL/KaLwRij mice resulted in the development of bone disease characterized by a significant decrease in cancellous bone volume in the tibial and femoral metaphyses, an increase in osteoclast formation, and radiologic evidence of osteolytic bone lesions. Dual-energy x-ray absorptiometry demonstrated a decrease in bone mineral density (BMD) at each of these sites. Treatment of mice with established myeloma with recombinant osteoprotegerin (OPG) protein, the soluble decoy receptor for RANKL, prevented the development of lytic bone lesions. OPG treatment was associated with preservation of cancellous bone volume and inhibition of osteoclast formation. OPG also promoted an increase in femoral, tibial, and vertebral BMD. These data suggest that the RANKL/RANK/OPG system may play a critical role in the development of osteolytic bone disease in multiple myeloma and that targeting this system may have therapeutic potential.


Asunto(s)
Glicoproteínas/uso terapéutico , Mieloma Múltiple/complicaciones , Osteólisis/prevención & control , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Animales , Densidad Ósea , Huesos/patología , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Citometría de Flujo , Expresión Génica , Glicoproteínas/administración & dosificación , Masculino , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Mieloma Múltiple/química , Mieloma Múltiple/patología , Trasplante de Neoplasias , Osteólisis/etiología , Osteólisis/patología , Osteoprotegerina , Ligando RANK , ARN Mensajero/análisis , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/administración & dosificación , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Clin Cancer Res ; 7(10): 2977-83, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11595685

RESUMEN

PURPOSE: Osteoprotegerin (OPG) is a soluble osteoclastogenesis inhibitor that regulates bone turnover. We reported recently that OPG protein expression is significantly increased in prostate cancer (CaP) cells present in bone metastases. The aim of this study was to determine serum OPG levels in patients at different stages of CaP and correlate the results with disease status. EXPERIMENTAL DESIGN: OPG levels were examined in patients with benign prostatic hyperplasia, clinically localized CaP, early recurrence of CaP, and advanced CaP and evidence of bone metastases. Serum OPG levels were measured by sandwich ELISA assays. The serum Crosslaps (sCTX) assay was used to quantify bone resorption in the advanced CaP group. RESULTS: Serum OPG levels were increased significantly in the advanced CaP group versus all other groups. There was no significant correlation between serum OPG levels and PSA levels either in the advanced CaP group or within any of three treatment subclasses of this group: no Tx, those not treated; Tx, those treated; and R, those treated with resorption blockers. Levels of OPG were negatively correlated with sCTX levels only in the advanced CaP Tx group. sCTX levels correlated with prostate-specific antigen levels in the advanced CaP Tx and R groups but not in the no-Tx group. CONCLUSIONS: Our data show that serum OPG levels are increased with advanced CaP. We hypothesize that OPG levels are related to CaP progression and suggest that further studies of the biological effects of OPG on CaP metastases are warranted.


Asunto(s)
Glicoproteínas/sangre , Neoplasias de la Próstata/patología , Receptores Citoplasmáticos y Nucleares/sangre , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Óseas/sangre , Neoplasias Óseas/secundario , Colágeno/sangre , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/patología , Osteoprotegerina , Fragmentos de Péptidos/sangre , Hiperplasia Prostática/sangre , Hiperplasia Prostática/patología , Neoplasias de la Próstata/sangre , Receptores del Factor de Necrosis Tumoral
12.
Endocrinology ; 142(10): 4295-304, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11564687

RESUMEN

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


Asunto(s)
Enfermedades Óseas Metabólicas/tratamiento farmacológico , Glicoproteínas/farmacología , Fragmentos de Péptidos/farmacología , Teriparatido/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Enfermedades Óseas Metabólicas/fisiopatología , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Glicoproteínas/uso terapéutico , Osteoprotegerina , Ovariectomía , Fragmentos de Péptidos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Receptores del Factor de Necrosis Tumoral , Teriparatido/análogos & derivados , Teriparatido/uso terapéutico
13.
J Orthop Res ; 19(4): 518-23, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11518255

RESUMEN

This study examines the ability of osteoprotegerin (OPG) to prevent the local bone resorption caused by sciatic nerve damage. Sixty-five 18-week-old male mice were assigned to one of six groups (n = 10-11/group). A baseline control group was sacrificed on day zero of the 10-day study. The remaining groups were placebo sham operated, placebo nerve crush (Plac NC) operated, 0.1 mg/kg/day OPG + nerve crush (LOW), 0.3 mg/kg/day OPG + nerve crush (MED), and 1.0 mg/kg/day OPG + nerve crush (HI). Nerve crush or sham operations were performed on the right leg. The left leg served as a contralateral control to the nerve crushed (ipsilateral) leg. The difference in mass between the right and left femur and tibia was examined. Additionally, quantitative histomorphometry was performed on the right and left femur and tibia diaphyses. Nerve crush resulted in a significant loss of bone mass in the ipsilateral side compared to the contralateral side. Bone mass for the ipsilateral bones of the Plac NC group were significantly reduced by 3.8% in the femur and 3.5% in the tibia compared to the contralateral limb. The percent diminution was reduced for OPG treated mice compared to the Plac NC group for both the femur and tibia. In the femur, the percent reduction of ipsilateral bone mass was reduced to 1.0% (LOW), 1.3% (MED) and 1.6% (HI) compared to the contralateral limb. In the tibia, loss of bone mass in the ipsilateral limb was reduced to 1.4% (LOW), 1.4% (MED), and 2.4% (HI) compared to the contralateral. OPG also decreased the amount of tibial endocortical resorption compared to the Plac NC group. In summary, OPG mitigated bone loss caused by damage to the sciatic nerve.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Glicoproteínas/farmacología , Nervio Ciático/fisiología , Animales , Resorción Ósea/patología , Diáfisis/patología , Modelos Animales de Enfermedad , Fémur/inervación , Fémur/patología , Húmero/anatomía & histología , Masculino , Ratones , Ratones Endogámicos C57BL , Compresión Nerviosa , Tamaño de los Órganos , Osteoprotegerina , Receptores Citoplasmáticos y Nucleares , Receptores del Factor de Necrosis Tumoral , Tibia/inervación , Tibia/patología
14.
Cancer Res ; 61(11): 4432-6, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11389072

RESUMEN

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


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/secundario , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Glicoproteínas/farmacología , Osteólisis/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Neoplasias Óseas/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Transformada , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Desnudos , Osteoprotegerina , Receptores Citoplasmáticos y Nucleares , Receptores del Factor de Necrosis Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Ther ; 3(2): 197-205, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11237676

RESUMEN

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


Asunto(s)
Adenoviridae/genética , Glicoproteínas/genética , Osteoporosis/terapia , Receptores Citoplasmáticos y Nucleares/genética , Animales , Bioensayo , Southern Blotting , Western Blotting , Densidad Ósea/efectos de los fármacos , Resorción Ósea , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Osteoprotegerina , Ovariectomía , Ovario/fisiología , Pelvis/diagnóstico por imagen , Radiografía , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo
16.
J Bone Miner Res ; 16(2): 348-60, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11204435

RESUMEN

Osteoprotegerin (OPG), a tumor necrosis factor (TNF) receptor family member, is a critical regulator of bone resorption. It is an important inhibitor of the terminal differentiation and activation of osteoclasts. This randomized, double-blind, placebo-controlled, sequential dose escalation study was conducted in postmenopausal women to determine the effect of a single subcutaneous (s.c.) dose of OPG on bone resorption as indicated by the biochemical markers, urinary N-telopeptide (NTX) and deoxypyridinoline (DPD), which are stable collagen degradation products. NTX levels decreased within 12 h after OPG administration. At the highest dose administered (3.0 mg/kg), a mean percent decrease in NTX of approximately 80% was observed 4 days after dosing. Six weeks after dosing a mean decrease of 14% in NTX was observed. The levels of bone-specific alkaline phosphatase (BSAP), a marker of bone formation, did not change for approximately 3 weeks after dosing. Thereafter, a modest decrease, reaching approximately 30% at 6 weeks, was observed in the 3.0-mg/kg dose group. The rapid decrease from baseline in NTX and delayed decrease in BSAP indicated that OPG acted primarily on osteoclasts to decrease bone resorption. OPG injections are well tolerated. This study, for the first time, indicates that a single s.c. injection of OPG is effective in rapidly and profoundly reducing bone turnover for a sustained period and that OPG therefore may be effective in treatment of bone diseases characterized by increased bone resorption such as osteoporosis.


Asunto(s)
Glicoproteínas/uso terapéutico , Osteoporosis/prevención & control , Posmenopausia , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Método Doble Ciego , Femenino , Glicoproteínas/administración & dosificación , Glicoproteínas/efectos adversos , Humanos , Persona de Mediana Edad , Osteoprotegerina , Placebos , Receptores Citoplasmáticos y Nucleares/administración & dosificación , Receptores del Factor de Necrosis Tumoral
17.
Biochem Biophys Res Commun ; 280(1): 334-9, 2001 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-11162519

RESUMEN

Osteoporosis and vasculopathy are common after organ transplantation and have been largely attributed to the use of immunosuppressants. Osteoprotegerin (OPG) is produced by osteoblastic and arterial cells, and inhibits osteoclast functions by neutralizing receptor activator of NF-kappaB ligand (RANKL). Because OPG-deficient mice develop osteoporosis and arterial calcification, we assessed the effects of immunosuppressants on OPG and RANKL expression by human osteoblastic and coronary artery smooth muscle cells (CASMC). Cyclosporine A, rapamycin, and FK-506 decreased OPG mRNA and protein levels in undifferentiated marrow stromal cells (by 63, 44, and 68%, respectively, P < 0.001). All three immunosuppressants increased RANKL mRNA levels in these cells by 60 to 210%. In contrast to these effects on marrow stromal cells, rapamycin, which may be relatively bone-sparing, increased OPG mRNA and protein production (by 120%, P < 0.001) in mature osteoblastic cells. Cyclosporine A also decreased OPG mRNA and protein production (by 52%, P < 0.001) of CASMC. In conclusion, immunosuppressants decrease OPG mRNA and protein production and increase RANKL gene expression by marrow stromal cells, and cyclosporine suppresses OPG production in CASMC. These studies thus provide a potential mechanism for immunosuppressant-induced bone loss, and the propensity of cyclosporine A to cause vascular disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Vasos Coronarios/metabolismo , Glicoproteínas/genética , Inmunosupresores/farmacología , Glicoproteínas de Membrana/metabolismo , Músculo Liso Vascular/citología , Osteoblastos/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Transcripción Genética/efectos de los fármacos , Células de la Médula Ósea/citología , Proteínas Portadoras/efectos de los fármacos , Línea Celular , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Ciclosporina/farmacología , Feto , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Glicoproteínas de Membrana/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , FN-kappa B/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoprotegerina , Ligando RANK , ARN Mensajero/genética , Receptor Activador del Factor Nuclear kappa-B , Receptores del Factor de Necrosis Tumoral/genética , Sirolimus/farmacología , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Tacrolimus/farmacología
18.
J Mater Sci Mater Med ; 12(7): 583-8, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15348250

RESUMEN

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

19.
Endocrinology ; 141(12): 4768-76, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11108292

RESUMEN

The one or more molecular mechanisms that determine the obligatory sequence of resorption followed by formation during bone remodeling is unclear. RANK ligand (RANK-L) is an essential requirement for osteoclastogenesis, and its activity is neutralized by binding to the soluble decoy receptor, osteoprotegerin (OPG). Because both molecules are produced by osteoblast lineage cells, we studied their developmental regulation in a conditionally immortalized human marrow stromal (hMS[2-15]) cell line. These cells can simulate the complete developmental sequence from undifferentiated precursor(s) to cells with the complete osteoblast phenotype that are capable of forming mineralized nodules. During osteoblast differentiation, RANK-L messenger RNA levels decreased by 5-fold, whereas OPG messenger RNA levels increased by 7-fold, resulting in a 35-fold change in the RANK-L/OPG ratio. OPG protein also increased by 6-fold. Mouse bone marrow cells generated osteoclast-like cells in coculture with undifferentiated hMS(2-15) cells, but did not when cocultured with hMS(2-15) cells in varying stages of differentiation, unless an excess of RANK-L was added. Thus, undifferentiated marrow stromal cells with a high RANK-L/OPG ratio can initiate and support osteoclastogenesis, but after differentiation to the mature osteoblast phenotype, they cannot. We speculate that the developmental regulation of OPG and RANK-L production by stromal/osteoblast cells contributes to the coordinated sequence of osteoclast and osteoblast differentiation during the bone remodeling cycle.


Asunto(s)
Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas de Membrana/genética , Osteoblastos/citología , Osteoclastos/citología , Receptores Citoplasmáticos y Nucleares/genética , Células del Estroma/citología , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Humanos , Cinética , Ratones , Osteoprotegerina , Ligando RANK , ARN Mensajero/análisis , Receptor Activador del Factor Nuclear kappa-B , Receptores del Factor de Necrosis Tumoral , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/fisiología
20.
J Exp Med ; 192(4): 463-74, 2000 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-10952716

RESUMEN

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


Asunto(s)
Densidad Ósea/fisiología , Calcinosis/fisiopatología , Glicoproteínas/metabolismo , Osteoclastos/metabolismo , Osteopetrosis/metabolismo , Osteoporosis/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Aorta/patología , Western Blotting , Células CHO , Catepsina K , Catepsinas/metabolismo , Cricetinae , Fémur/anatomía & histología , Fémur/diagnóstico por imagen , Fémur/metabolismo , Glicoproteínas/genética , Humanos , Inmunohistoquímica , Hibridación in Situ , Isoenzimas/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/ultraestructura , Osteopetrosis/genética , Osteoporosis/genética , Osteoprotegerina , Radiografía , Receptores Citoplasmáticos y Nucleares/genética , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes de Fusión/metabolismo , Fosfatasa Ácida Tartratorresistente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...