Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Expert Opin Drug Saf ; 23(9): 1069-1077, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39262109

RESUMEN

INTRODUCTION: Denosumab (Prolia) is a fully human monoclonal antibody against the receptor activator of the nuclear factor kappaB ligand. It is a potent antiresorptive agent that reduces osteoclastogenesis. AREAS COVERED: Denosumab has been shown to improve bone mineral density and reduce the incidence of new fractures in postmenopausal women and men. It is also used in the treatment of glucocorticoid-induced osteoporosis, as well as for the prevention of bone loss and reduction of fracture risk in men receiving androgen deprivation therapy for non-metastatic prostate cancer and women receiving adjuvant aromatase inhibitor therapy for breast cancer. Initial safety concerns included infections, cancer, skin reactions, cardiovascular disease, hypocalcemia, osteonecrosis of the jaw, and atypical femur fractures; however, further study and experience provide reassurance on these issues. Anecdotal reports have raised concerns about an increased risk of multiple vertebral fractures following discontinuation of denosumab. EXPERT OPINION: Although bisphosphonates are often selected as initial therapy for osteoporosis, denosumab may be an appropriate initial therapy in patients at high risk for fracture, including older patients who have difficulty with the dosing requirements of oral bisphosphonates, as well as patients who are intolerant of, unresponsive to, or have contraindications to other therapies. Additional data is needed to address questions regarding treatment duration and discontinuation.


Asunto(s)
Conservadores de la Densidad Ósea , Denosumab , Osteoporosis , Humanos , Denosumab/efectos adversos , Denosumab/administración & dosificación , Osteoporosis/inducido químicamente , Osteoporosis/tratamiento farmacológico , Conservadores de la Densidad Ósea/efectos adversos , Conservadores de la Densidad Ósea/administración & dosificación , Femenino , Densidad Ósea/efectos de los fármacos , Masculino , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacología , Ligando RANK/efectos adversos , Ligando RANK/antagonistas & inhibidores , Ligando RANK/administración & dosificación , Animales , Difosfonatos/efectos adversos , Difosfonatos/administración & dosificación , Fracturas Óseas/inducido químicamente , Fracturas Óseas/prevención & control
2.
Int Immunopharmacol ; 88: 106872, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32769069

RESUMEN

Receptor activator of NF-κB ligand (RANKL) as an osteoclast differentiation factor induces inflammatory reactions via production of thymic stromal lymphopoietin (TSLP). Epigallocatechin gallate (EGCG) is the major and the most active compound in green tea and has anti-inflammatory, anti-cancer, anti-oxidant, and neuroprotective effects. However, the effect and molecular mechanisms of EGCG are still unknown in RANKL-induced inflammatory reactions. Here we investigated the immuno-regulatory effects and its molecular mechanisms of epigallocatechin gallate (EGCG) in RANKL-stimulated human mast cell line, HMC-1 cells. In this study, EGCG prevented expression of PI3 Kinase and phosphorylation of mitogen-activated protein (MAP) Kinases in RANKL-stimulated HMC-1 cells. EGCG prevented caspase-1 activity and decreased transcriptional activity of nuclear factor (NF)-κB by suppressing inhibitory protein κBα phosphorylation in RANKL-stimulated HMC-1 cells. EGCG has been shown to prevent production and mRNA expression of TSLP, interleukin (IL)-1ß, IL-6, and IL-8 by RANKL without cytotoxicity. Furthermore, EGCG prevented degranulation of mast cell in RANKL-stimulated HMC-1 cells. Overall, these results suggest that EGCG acts as a natural agent for preventing and treating RANKL-mediated inflammatory diseases by targeting PI3 Kinase, MAP Kinase, caspase-1, and NF-κB signaling cascade in mast cells.


Asunto(s)
Catequina/análogos & derivados , Inflamación/metabolismo , Mastocitos/efectos de los fármacos , Ligando RANK/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Caspasa 1/efectos de los fármacos , Caspasa 1/metabolismo , Catequina/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Elafina/efectos de los fármacos , Elafina/metabolismo , Histamina/metabolismo , Humanos , Inflamación/inducido químicamente , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucina-8/efectos de los fármacos , Interleucina-8/metabolismo , Mastocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Ligando RANK/efectos adversos , Linfopoyetina del Estroma Tímico
3.
Mol Cell Biochem ; 464(1-2): 111-117, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31724123

RESUMEN

Numerous studies have highlighted the causal link between over-production of reactive oxygen species (ROS) and cardiovascular complications such as vascular calcification (VC). Receptor-activator of nuclear factor-κB ligand (RANKL) has previously been shown to act on endothelial cells, eliciting the production/release of paracrine pro-calcific signals that act, in-turn, upon underlying vascular smooth muscle cells (VSMCs) to induce osteoblastic differentiation and VC. A role for endothelial ROS signaling in this process has not been established however. In the current paper, we investigate the possibility that RANKL leads to ROS signaling within the endothelial layer as part of the RANKL-driven VC signaling cascade. Human aortic endothelial cells (HAECs) were treated with RANKL (25 ng/ml, 72 h) and monitored for ROS production, in parallel with various pro-calcific signaling indices. Antioxidant co-treatments included TRAIL (5 ng/ml), apocynin (10 mM) and N-acetylcysteine (5 mM). Treatment of HAECs with RANKL-induced robust ROS production. This surge could be partially attenuated by TRAIL and strongly attenuated by apocynin and N-acetylcysteine. RANKL also elicited a range of signaling events in HAECs that we have previously demonstrated are coupled to osteoblastic differentiation in underlying VSMCs. These include non-canonical NF-κB/p52 activation, elevated BMP-2 release and increased alkaline phosphatase (ALP) enzyme activity (cellular and extracellular). Importantly, these RANKL-induced signaling events could be completely prevented by co-treatment of HAECs with antioxidants. In summary, RANKL elicits ROS generation in HAECs with direct consequences for generation of paracrine pro-calcific signals known to effect calcification in underlying VSMCs.


Asunto(s)
Aorta/metabolismo , Células Endoteliales/metabolismo , Comunicación Paracrina/efectos de los fármacos , Ligando RANK/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/metabolismo , Adulto , Aorta/patología , Células Endoteliales/patología , Humanos , Masculino , Ligando RANK/farmacología , Calcificación Vascular/inducido químicamente , Calcificación Vascular/patología
4.
Tissue Eng Regen Med ; 16(3): 265-273, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31205855

RESUMEN

Background: Wear debris-induced osteolysis leads to periprosthetic loosening and subsequent prosthetic failure. Since excessive osteoclast formation is closely implicated in periprosthetic osteolysis, identification of agents to suppress osteoclast formation and/or function is crucial for the treatment and prevention of wear particle-induced bone destruction. In this study, we examined the potential effect of pentamidine treatment on titanium (Ti) particle-induced osteolysis, and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis. Methods: The effect of pentamidine treatment on bone destruction was examined in Ti particle-induced osteolysis mouse model. Ti particles were implanted onto mouse calvaria, and vehicle or pentamidine was administered for 10 days. Then, calvarial bone tissue was analyzed using micro-computed tomography and histology. We performed in vitro osteoclastogenesis assay using bone marrow-derived macrophages (BMMs) to determine the effect of pentamidine on osteoclast formation. BMMs were treated with 20 ng/mL RANKL and 10 ng/mL macrophage colony-stimulating factor in the presence or absence of pentamidine. Osteoclast differentiation was determined by tartrate-resistant acid phosphatase staining, real-time polymerase chain reaction, and immunofluorescence staining. Results: Pentamidine administration decreased Ti particle-induced osteoclast formation significantly and prevented bone destruction compared to the Ti particle group in vivo. Pentamidine also suppressed RANKL-induced osteoclast differentiation and actin ring formation markedly, and inhibited the expression of nuclear factor of activated T cell c1 and osteoclast-specific genes in vitro. Additionally, pentamidine also attenuated RANKL-mediated phosphorylation of IκBα in BMMs. Conclusion: These results indicate that pentamidine is effective in inhibiting osteoclast formation and significantly attenuates wear debris-induced bone loss in mice.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteólisis/tratamiento farmacológico , Pentamidina/farmacología , Ligando RANK/efectos adversos , Titanio/efectos adversos , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Osteólisis/inducido químicamente , Pentamidina/uso terapéutico , Prótesis e Implantes , Cráneo/metabolismo , Cráneo/patología , Microtomografía por Rayos X
5.
Actual. osteol ; 15(1): 34-43, ene. abr. 2019. ilus.
Artículo en Español | LILACS | ID: biblio-1049002

RESUMEN

La brucelosis es una de las enfermedades zoonóticas más importantes a nivel mundial capaz de producir enfermedad crónica en los seres humanos. La localización osteoarticular es la presentación más común de la enfermedad activa en el hombre. Sin embargo, algunos de los mecanismos moleculares implicados en la enfermedad osteoarticular han comenzado a dilucidarse recientemente. Brucella abortus induce daño óseo a través de diversos mecanismos en los cuales están implicados TNF-α y RANKL. En estos procesos participan células inflamatorias que incluyen monocitos/macrófagos, neutrófilos, linfocitos T del tipo Th17 y linfocitos B. Además, B. abortus puede afectar directamente las células osteoarticulares. La bacteria inhibe la deposición de la matriz ósea por los osteoblastos y modifica el fenotipo de estas células para producir metaloproteinasas de matriz (MMPs) y la secreción de citoquinas que contribuyen a la degradación del hueso. Por otro lado, la infección por B. abortus induce un aumento en la osteoclastogénesis, lo que aumenta la resorción de la matriz ósea orgánica y mineral y contribuye al daño óseo. Dado que la patología inducida por Brucella afecta el tejido articular, se estudió el efecto de la infección sobre los sinoviocitos. Estos estudios revelaron que, además de inducir la activación de estas células para secretar quemoquinas, citoquinas proinflamatorias y MMPs, la infección inhibe la muerte por apoptosis de los sinoviocitos. Brucella es una bacteria intracelular que se replica en el retículo endoplásmico de los macrófagos. El análisis de los sinoviocitos infectados con B. abortus indicó que las bacterias también se multiplican en el retículo endoplasmático, lo que sugiere que la bacteria podría usar este tipo celular para la multiplicación intracelular durante la localización osteoarticular de la enfermedad. Los hallazgos presentados en esta revisión intentan responder a preguntas sobre los mediadores inflamatorios implicados en el daño osteoarticular causado por Brucella. (AU)


Brucellosis is one of the most important zoonotic diseases that can produce chronic disease in humans worldwide. Osteoarticular involvement is the most common presentation of human active disease. The molecular mechanisms implicated in bone damage have started to be elucidated. B. abortus induces bone damage through diverse mechanisms in which TNF-α and RANKL are implicated. These processes are driven by inflammatory cells, including monocytes/macrophages, neutrophils, Th17 lymphocytes and B cells. Also, Brucella abortus (B. abortus) can directly affect osteoarticular cells. The bacterium inhibits bone matrix deposition by osteoblast and modifies the phenotype of these cells to produce matrix methalloproteinases (MMPs) and cytokine secretion that contribute to bone matrix degradation. B. abortus also affects osteoclast increasing mineral and organic bone matrix resorption and contributing to bone damage. Since the pathology induced by Brucella species involves joint tissue, experiments conducted in sinoviocytes revealed that besides inducing the activation of these cells to secrete chemokines, proinflammatory cytokines and MMPS, the infection also inhibits sinoviocyte apoptosis. Brucella is an intracellular bacterium that replicate in the endoplasmic reticulum of macrophages. The analysis of B. abortus infected sinoviocytes indicated that bacteria also replicate in their reticulum suggesting that the bacterium could use this cell type for intracellular replication during the osteoarticular localization of the disease. The findings presented in this review try to answer key questions about the inflammatory mediators involved in osteoarticular damage caused by Brucella. (AU)


Asunto(s)
Humanos , Animales , Osteoartritis/patología , Brucella abortus/patogenicidad , Brucelosis/patología , Osteoartritis/inmunología , Osteoblastos/patología , Osteocitos/microbiología , Osteogénesis/inmunología , Brucella abortus/inmunología , Brucelosis/etiología , Brucelosis/inmunología , Linfocitos B/patología , Citocinas/efectos adversos , Factor de Necrosis Tumoral alfa/efectos adversos , Metaloproteinasas de la Matriz/síntesis química , Ligando RANK/efectos adversos , Células Th17/patología , Sinoviocitos/inmunología , Macrófagos/patología , Neutrófilos/patología
6.
Cancer Lett ; 443: 135-144, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30540926

RESUMEN

Bone metastasis causes bone pain and pathological bone fracture in breast cancer patients with a serious complication. Previous studies have demonstrated that a novel phosphatidyl inositol 3-kinase (PI3K)-mTOR inhibitor PKI-402 suppressed the growth of breast cancer cells. However, the role of PKI-402 involved in osteolysis induced by breast cancer remains unclear. In this study, we showed that treatment of PKI-402 led to significant decreases in RANKL-induced osteoclastogenesis and osteoclast-specific gene expression in mouse bone marrow-derived macrophages and reduced proliferation, migration and invasion of MDA-MB-231 breast cancer cells by blocking the PI3K-AKT-mTOR signaling pathway. Importantly, as evidenced by the observation that the administration of PKI-402 inhibited MDA-MB-231-induced osteolysis in vivo, PKI-402 exerted an inhibitory effect on osteoclast formation and bone resorption, critical for cancer cells-induced bone destruction. These results strongly suggest that PKI-402 might have a therapeutic potential to inhibit breast cancer induced osteolysis.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Osteólisis/prevención & control , Compuestos de Fenilurea/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/administración & dosificación , Ligando RANK/efectos adversos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Osteogénesis/efectos de los fármacos , Osteólisis/genética , Osteólisis/metabolismo , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Am J Chin Med ; 45(8): 1725-1744, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29121799

RESUMEN

Puerariae radix, the dried root of Pueraria lobate Ohwi, is known to prevent bone loss in ovariectomized mice; however, the precise molecular mechanisms are not understood. In this study, we investigated the effects and underlying mechanisms of action of Puerariae radix extract (PRE) on receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis. PRE dose-dependently inhibited osteoclast differentiation and formation, decreased the bone-resorbing activity of osteoclasts, and downregulated the expression of osteoclast differentiation marker genes. The expression of osteoclastogenic factors produced by PRE-treated osteoblasts such as RANKL, macrophage colony-stimulating factor (M-CSF), and osteoprotegerin (OPG) was comparable to that of untreated (control) cells. However, the formation of osteoclasts via bone marrow cell and calvaria-derived osteoblast co-cultures was suppressed by PRE treatment. Therefore, the inhibitory effects of PRE on osteoclastogenesis clearly targeted osteoclasts, but not osteoblasts. PRE treatment considerably reduced RANKL-induced mitogen-activated protein kinases (MAPKs) activity, especially c-Jun N-terminal kinase, in osteoclast precursor cells. In addition, PRE markedly suppressed cAMP response element-binding protein (CREB) activation and the induction of peroxisome proliferator-activated receptor gamma coactivator 1ß (PGC1ß), which stimulate osteoclastogenesis - an effect that was not observed for puerarin and 17-ß estradiol. Finally, PRE treatment significantly repressed the expression of c-Fos and the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), which is a master transcription factor for osteoclastogenesis in vitro and in vivo. Overall, these results strongly suggest that PRE is an effective inhibitor of RANKL-induced osteoclastogenesis and may be a potent therapeutic agent for bone-related diseases such as osteoporosis, rheumatoid arthritis, and periodontitis.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Extractos Vegetales/farmacología , Pueraria/química , Ligando RANK/efectos adversos , Transducción de Señal/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citocinas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Fitoterapia , Extractos Vegetales/aislamiento & purificación , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Nutrients ; 9(5)2017 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-28452958

RESUMEN

Osteoclasts are large, multinucleated cells that are responsible for the breakdown or resorption of bone during bone remodelling. Studies have shown that certain fatty acids (FAs) can increase bone formation, reduce bone loss, and influence total bone mass. Palmitoleic acid (PLA) is a 16-carbon, monounsaturated FA that has shown anti-inflammatory properties similar to other FAs. The effects of PLA in bone remain unexplored. Here we investigated the effects of PLA on receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL)-induced osteoclast formation and bone resorption in RAW264.7 murine macrophages. PLA decreased the number of large, multinucleated tartrate resistant acid phosphatase (TRAP) positive osteoclasts and furthermore, suppressed the osteolytic capability of these osteoclasts. This was accompanied by a decrease in expression of resorption markers (Trap, matrix metalloproteinase 9 (Mmp9), cathepsin K (Ctsk)). PLA further decreased the expression of genes involved in the formation and function of osteoclasts. Additionally, PLA inhibited NF-κB activity and the activation of mitogen activated protein kinases (MAPK), c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK). Moreover, PLA induced apoptosis in mature osteoclasts. This study reveals that PLA inhibits RANKL-induced osteoclast formation in RAW264.7 murine macrophages through suppression of NF-κB and MAPK signalling pathways. This may indicate that PLA has potential as a therapeutic for bone diseases characterized by excessive osteoclast formation.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Ácidos Grasos Monoinsaturados/farmacología , Sistema de Señalización de MAP Quinasas , FN-kappa B/genética , Osteogénesis/efectos de los fármacos , Ligando RANK/efectos adversos , Animales , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Resorción Ósea/etiología , Catepsina K/genética , Catepsina K/metabolismo , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Células RAW 264.7 , Transducción de Señal , Fosfatasa Ácida Tartratorresistente/genética , Fosfatasa Ácida Tartratorresistente/metabolismo
9.
Sci Rep ; 6: 30918, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27480204

RESUMEN

Bone homeostasis is maintained by the sophisticated coupled actions of bone-resorbing osteoclasts and bone-forming osteoblasts. Here we identify activating transcription factor 3 (ATF3) as a pivotal transcription factor for the regulation of bone resorption and bone remodeling under a pathological condition through modulating the proliferation of osteoclast precursors. The osteoclast precursor-specific deletion of ATF3 in mice led to the prevention of receptor activator of nuclear factor-κB (RANK) ligand (RANKL)-induced bone resorption and bone loss, although neither bone volume nor osteoclastic parameter were markedly altered in these knockout mice under the physiological condition. RANKL-dependent osteoclastogenesis was impaired in vitro in ATF3-deleted bone marrow macrophages (BMM). Mechanistically, the deficiency of ATF3 impaired the RANKL-induced transient increase in cell proliferation of osteoclast precursors in bone marrow in vivo as well as of BMM in vitro. Moreover, ATF3 regulated cyclin D1 mRNA expression though modulating activator protein-1-dependent transcription in the osteoclast precursor, and the introduction of cyclin D1 significantly rescued the impairment of osteoclastogenesis in ATF3-deleted BMM. Therefore, these findings suggest that ATF3 could have a pivotal role in osteoclastogenesis and bone homeostasis though modulating cell proliferation under pathological conditions, thereby providing a target for bone diseases.


Asunto(s)
Factor de Transcripción Activador 3/fisiología , Remodelación Ósea , Resorción Ósea/prevención & control , Osteoclastos/citología , Ligando RANK/efectos adversos , Animales , Células de la Médula Ósea/metabolismo , Resorción Ósea/etiología , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Proliferación Celular , Regulación de la Expresión Génica , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/metabolismo
10.
J Immunol ; 193(3): 1383-91, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24958900

RESUMEN

The glycoprotein milk fat globule-epidermal growth factor factor 8 (MFG-E8) is expressed in several tissues and mediates diverse homeostatic functions. However, whether it plays a role in bone homeostasis has not been established. In this study, we show for the first time, to our knowledge, that osteoclasts express and are regulated by MFG-E8. Bone marrow-derived osteoclast precursors from MFG-E8-deficient (Mfge8(-/-)) mice underwent increased receptor activator of NF-κB ligand-induced osteoclastogenesis, leading to enhanced resorption pit formation compared with wild-type controls. Consistently, exogenously added MFG-E8 inhibited receptor activator of NF-κB ligand-induced osteoclastogenesis from mouse or human osteoclast precursors. Upon induction of experimental periodontitis, an oral inflammatory disease characterized by loss of bone support of the dentition, Mfge8(-/-) mice exhibited higher numbers of osteoclasts and more bone loss than did wild-type controls. Accordingly, local microinjection of anti-MFG-E8 mAb exacerbated periodontal bone loss in wild-type mice. Conversely, microinjection of MFG-E8 inhibited bone loss in experimental mouse periodontitis. In comparison with wild-type controls, Mfge8(-/-) mice also experienced >60% more naturally occurring chronic periodontal bone loss. In conclusion, MFG-E8 is a novel homeostatic regulator of osteoclasts that could be exploited therapeutically to treat periodontitis and perhaps other immunological disorders associated with inflammatory bone loss.


Asunto(s)
Antígenos de Superficie/fisiología , Resorción Ósea/inmunología , Homeostasis/inmunología , Mediadores de Inflamación/fisiología , Osteoclastos/inmunología , Osteoclastos/patología , Animales , Antígenos de Superficie/biosíntesis , Antígenos de Superficie/genética , Resorción Ósea/genética , Resorción Ósea/patología , Línea Celular , Homeostasis/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Leche/biosíntesis , Proteínas de la Leche/genética , Periodontitis/genética , Periodontitis/inmunología , Periodontitis/patología , Ligando RANK/administración & dosificación , Ligando RANK/efectos adversos , Células Madre/inmunología , Células Madre/metabolismo , Células Madre/patología
11.
Ugeskr Laeger ; 174(47): 2926-30, 2012 Nov 19.
Artículo en Danés | MEDLINE | ID: mdl-23171789

RESUMEN

Both chronic kidney disease and osteoporosis are frequent conditions in the general population. Most drugs for treating osteoporosis seem safe in terms of affecting renal function for patients with mildly to moderate decreased renal function. There are very few data on the efficacy (reduction in fracture risk) or safety in patients with severely decreased renal function (glomerular filtration rate < 30 ml/min) or on dialysis.


Asunto(s)
Osteoporosis/tratamiento farmacológico , Insuficiencia Renal/complicaciones , Anciano , Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/efectos adversos , Conservadores de la Densidad Ósea/farmacocinética , Calcio/metabolismo , Difosfonatos/administración & dosificación , Difosfonatos/efectos adversos , Difosfonatos/farmacocinética , Femenino , Fracturas Espontáneas/diagnóstico por imagen , Tasa de Filtración Glomerular , Humanos , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Tasa de Depuración Metabólica , Osteoporosis/complicaciones , Osteoporosis/metabolismo , Fracturas Osteoporóticas/diagnóstico por imagen , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/efectos adversos , Hormona Paratiroidea/farmacocinética , Ligando RANK/administración & dosificación , Ligando RANK/efectos adversos , Ligando RANK/farmacocinética , Radiografía , Diálisis Renal , Insuficiencia Renal/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Moduladores Selectivos de los Receptores de Estrógeno/efectos adversos , Moduladores Selectivos de los Receptores de Estrógeno/farmacocinética , Estroncio/administración & dosificación , Estroncio/efectos adversos , Estroncio/farmacocinética
12.
J Bone Miner Res ; 27(12): 2501-10, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22836362

RESUMEN

In the last decades the molecular basis of monogenic diseases has been largely unraveled, although their treatment has often remained unsatisfactory. Autosomal recessive osteopetrosis (ARO) belongs to the small group of genetic diseases that are usually treated with hematopoietic stem cell transplantation (HSCT). However, this approach is not effective in the recently identified form carrying mutations in the receptor activator of NF-κB ligand (RANKL) gene. In this subset, therapy replacement approach based on RANKL delivery has a strong rationale. Here we demonstrate that the systematic administration of RANKL for 1 month to Rankl(-/-) mice, which closely resemble the human disease, significantly improves the bone phenotype and has beneficial effects on bone marrow, spleen and thymus; major adverse effects arise only when mice are clearly overtreated. Overall, we provide evidence that the pharmacological administration of RANKL represents the appropriate treatment option for RANKL-deficient ARO patients, to be validated in a pilot clinical trial.


Asunto(s)
Osteopetrosis/tratamiento farmacológico , Osteopetrosis/genética , Ligando RANK/uso terapéutico , Animales , Células de la Médula Ósea/efectos de los fármacos , Resorción Ósea/inducido químicamente , Huesos/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Osteopetrosis/patología , Fenotipo , Ligando RANK/administración & dosificación , Ligando RANK/efectos adversos , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/deficiencia , Receptor Activador del Factor Nuclear kappa-B/genética
13.
Ann N Y Acad Sci ; 1263: 29-40, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22831177

RESUMEN

Denosumab is a fully human monoclonal antibody against RANK ligand (RANKL), an essential cytokine for the formation, function, and survival of osteoclasts. The role of excessive RANKL as a contributor to conditions characterized by bone loss or bone destruction has been well studied. With its novel mechanism of action, denosumab offers a significant advance in the treatment of postmenopausal osteoporosis; bone loss associated with hormone ablation therapy in women with breast cancer and men with prostate cancer; and the prevention of skeletal-related events in patients with bone metastases from solid tumors by offering clinical benefit to these patients in need.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Descubrimiento de Drogas , Ligando RANK/antagonistas & inhibidores , Animales , Antineoplásicos Hormonales/efectos adversos , Neoplasias Óseas/epidemiología , Neoplasias Óseas/inmunología , Resorción Ósea/epidemiología , Resorción Ósea/etiología , Resorción Ósea/inmunología , Denosumab , Humanos , Osteoporosis/epidemiología , Osteoporosis/etiología , Osteoporosis/inmunología , Ligando RANK/efectos adversos , Resultado del Tratamiento
14.
Lancet ; 379(9810): 39-46, 2012 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-22093187

RESUMEN

BACKGROUND: Bone metastases are a major cause of morbidity and mortality in men with prostate cancer. Preclinical studies suggest that osteoclast inhibition might prevent bone metastases. We assessed denosumab, a fully human anti-RANKL monoclonal antibody, for prevention of bone metastasis or death in non-metastatic castration-resistant prostate cancer. METHODS: In this phase 3, double-blind, randomised, placebo-controlled study, men with non-metastatic castration-resistant prostate cancer at high risk of bone metastasis (prostate-specific antigen [PSA] ≥8·0 µg/L or PSA doubling time ≤10·0 months, or both) were enrolled at 319 centres from 30 countries. Patients were randomly assigned (1:1) via an interactive voice response system to receive subcutaneous denosumab 120 mg or subcutaneous placebo every 4 weeks. Randomisation was stratified by PSA eligibility criteria and previous or ongoing chemotherapy for prostate cancer. Patients, investigators, and all people involved in study conduct were masked to treatment allocation. The primary endpoint was bone-metastasis-free survival, a composite endpoint determined by time to first occurrence of bone metastasis (symptomatic or asymptomatic) or death from any cause. Efficacy analysis was by intention to treat. The masked treatment phase of the trial has been completed. This trial was registered at ClinicalTrials.gov, number NCT00286091. FINDINGS: 1432 patients were randomly assigned to treatment groups (716 denosumab, 716 placebo). Denosumab significantly increased bone-metastasis-free survival by a median of 4·2 months compared with placebo (median 29·5 [95% CI 25·4-33·3] vs 25·2 [22·2-29·5] months; hazard ratio [HR] 0·85, 95% CI 0·73-0·98, p=0·028). Denosumab also significantly delayed time to first bone metastasis (33·2 [95% CI 29·5-38·0] vs 29·5 [22·4-33·1] months; HR 0·84, 95% CI 0·71-0·98, p=0·032). Overall survival did not differ between groups (denosumab, 43·9 [95% CI 40·1-not estimable] months vs placebo, 44·8 [40·1-not estimable] months; HR 1·01, 95% CI 0·85-1·20, p=0·91). Rates of adverse events and serious adverse events were similar in both groups, except for osteonecrosis of the jaw and hypocalcaemia. 33 (5%) patients on denosumab developed osteonecrosis of the jaw versus none on placebo. Hypocalcaemia occurred in 12 (2%) patients on denosumab and two (<1%) on placebo. INTERPRETATION: This large randomised study shows that targeting of the bone microenvironment can delay bone metastasis in men with prostate cancer. FUNDING: Amgen Inc.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias Óseas/secundario , Orquiectomía , Neoplasias de la Próstata/tratamiento farmacológico , Ligando RANK/administración & dosificación , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Denosumab , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Método Doble Ciego , Humanos , Inyecciones Subcutáneas , Masculino , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Ligando RANK/efectos adversos
16.
Tidsskr Nor Laegeforen ; 131(19): 1893-6, 2011 Oct 04.
Artículo en Noruego | MEDLINE | ID: mdl-21984295

RESUMEN

BACKGROUND: Treatment with bisphosphonates reduces the risk of new fractures and is the treatment of choice for osteoporosis. Denosumab inhibits bone resorption via a different mechanism than bisphosphonates, and is a new option in the treatment of osteoporosis. In this paper we give an overview of the mode of action and clinical effects. MATERIAL AND METHODS: The paper is based on a non-systematic literature search in Pubmed/Medline. RESULTS: Denosumab is a human monoclonal antibody to receptor-activated nuclear factor kappa B (RANKL), a member of the TNF family that is formed in the osteoblast. Binding to RANKL results in reduced recruitment and activity of osteoclasts. Denosumab 60 mg given subcutaneously every six months is shown to inhibit bone resorption to a greater degree than bisphosphonates. In a three-year study of 7,868 women with postmenopausal osteoporosis, a reduction in the relative risk of vertebral, non-vertebral and hip fractures compared to placebo was found (68. 20 and 40 %, correspondingly). In the clinical trials with denosumab, the safety profile was similar to placebo, except for a slightly higher incidence of cellulitis and exanthema. Denosumab has also shown promising skeletal effects in the treatment of cancer and rheumatoid arthritis. INTERPRETATION: Treatment with denosumab has an effect on postmenopausal osteoporosis and may be an alternative to treatment with bisphosphonates. There are few adverse effects and it is simple to administer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Osteoporosis Posmenopáusica/tratamiento farmacológico , Ligando RANK/uso terapéutico , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Densidad Ósea/efectos de los fármacos , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/prevención & control , Denosumab , Femenino , Humanos , Inyecciones Subcutáneas , Persona de Mediana Edad , Fracturas Osteoporóticas/prevención & control , Ligando RANK/administración & dosificación , Ligando RANK/efectos adversos
17.
Pharmacotherapy ; 31(5): 510-23, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21923432

RESUMEN

Osteoporosis is a degenerative bone disease affecting approximately 10 million American adults. Several options are available to prevent development of the disease or slow and even stop its progression. Nonpharmacologic measures include adequate intake of calcium and vitamin D, exercise, fall prevention, and avoidance of tobacco and excessive alcohol intake. Current drug therapy includes bisphosphonates, calcitonin, estrogen or hormone therapy, selective estrogen receptor modulators, and teriparatide. Denosumab, a receptor activator of nuclear factor-K B ligand (RANKL) inhibitor, was recently approved by the United States Food and Drug Administration for treatment of postmenopausal osteoporosis. Patients treated with denosumab experienced significant gains in bone mineral density, rapid reductions in markers of bone turnover, and a reduced risk for new vertebral fracture. Compared with placebo, patients receiving denosumab 60 mg subcutaneously once every 6 months experienced gains in bone mineral density of 6.5-11% when treated for 24-48 months. One trial demonstrated the superiority of denosumab compared with alendronate, but the differences were small. The most common adverse reactions to denosumab include back pain, pain in extremities, musculoskeletal pain, and cystitis. Serious, but rare, adverse reactions include the development of serious infections, dermatologic changes, and hypocalcemia. The recommended dosing of denosumab is 60 mg every 6 months as a subcutaneous injection in the upper arm, upper thigh, or abdomen. Although beneficial effects on bone mineral density and fracture rate have been established in clinical trials, the risks associated with denosumab must be evaluated before therapy initiation. Of concern is the risk of infection, and denosumab should likely be avoided in patients taking immunosuppressive therapy or at high risk for infection. Therefore, bisphosphonates will likely remain as first-line therapy. Denosumab should be considered in patients unable to tolerate or who have adherence issues or contraindications to other therapies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Densidad Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Osteoporosis Posmenopáusica/tratamiento farmacológico , Ligando RANK/uso terapéutico , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Biomarcadores/sangre , Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/efectos adversos , Conservadores de la Densidad Ósea/farmacocinética , Remodelación Ósea/efectos de los fármacos , Huesos/metabolismo , Denosumab , Femenino , Fracturas Óseas/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Osteoporosis/sangre , Osteoporosis/tratamiento farmacológico , Osteoporosis/fisiopatología , Osteoporosis Posmenopáusica/sangre , Osteoporosis Posmenopáusica/fisiopatología , Ligando RANK/administración & dosificación , Ligando RANK/efectos adversos , Ligando RANK/antagonistas & inhibidores , Ligando RANK/farmacocinética
18.
Drugs Today (Barc) ; 47(8): 605-13, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21850283

RESUMEN

Denosumab is a fully human monoclonal antibody that inhibits the formation, function and survival of osteoclasts, preventing the interaction of tumor necrosis factor ligand superfamily member 11 (receptor activator of nuclear factor kappa-B ligand, RANKL) with the tumor necrosis factor receptor superfamily member 11A (osteoclast differentiation factor receptor, ODFR, receptor activator of NF-KB, RANK). This results in a reduction in bone resorption and an increase in bone mineral density. In clinical studies, denosumab has been shown to decrease the risk for vertebral, hip and nonvertebral fractures in women with postmenopausal osteoporosis and the risk for new vertebral fractures in men with nonmetastatic prostate cancer receiving androgen deprivation therapy, with a rate of side effects similar to placebo. A number of clinical trials with denosumab are ongoing to demonstrate its value for other indications and to further characterize its effects on immunomodulation. Denosumab is a new alternative for the prevention and treatment of postmenopausal osteoporosis and a promising agent for the treatment of other bone diseases associated with bone loss.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Ligando RANK/uso terapéutico , Alendronato/uso terapéutico , Antagonistas de Andrógenos/efectos adversos , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Inhibidores de la Aromatasa/efectos adversos , Artritis Reumatoide/tratamiento farmacológico , Densidad Ósea/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Ensayos Clínicos como Asunto , Denosumab , Femenino , Humanos , Masculino , Osteoporosis Posmenopáusica/tratamiento farmacológico , Ligando RANK/efectos adversos
19.
Am J Health Syst Pharm ; 68(15): 1409-18, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21785030

RESUMEN

PURPOSE: The pharmacologic properties, clinical efficacy, and safety profile of the injectable agent denosumab for the treatment of postmenopausal women with osteoporosis are reviewed. SUMMARY: Denosumab, a human monoclonal antibody that targets a key protein mediator of bone resorption, was approved by the Food and Drug Administration in June 2010 for the treatment of postmenopausal women with osteoporosis who are at high risk for fracture, including "patients who have failed or are intolerant to other available osteoporosis therapy." Available in a 60-mg prefilled syringe, denosumab should be administered subcutaneously by a health care professional at six-month intervals. In Phase III clinical efficacy trials involving nearly 10,000 postmenopausal women, the use of denosumab was associated with a number of significant benefits: reduced bone resorption, increased bone mass, and reduced rates of vertebral, nonvertebral, and hip fractures. Results of two comparison studies indicated that denosumab therapy increased bone mineral density (BMD) at various skeletal sites to a significantly greater extent than alendronate therapy. In the largest clinical trial of the drug to date, adverse effects occurring significantly more often with denosumab versus placebo included eczema-related effects and cellulitis; long-term safety evaluations are ongoing. CONCLUSION: Denosumab has been shown to decrease bone resorption; increase BMD at all skeletal sites measured; and significantly reduce rates of vertebral, nonvertebral, and hip fractures in postmenopausal women with osteoporosis. Denosumab appears to have a favorable risk:benefit profile and provides a new treatment option for many patients in this population.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Osteoporosis Posmenopáusica/tratamiento farmacológico , Ligando RANK/uso terapéutico , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/efectos adversos , Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/tratamiento farmacológico , Denosumab , Femenino , Fracturas Óseas/etiología , Fracturas Óseas/prevención & control , Humanos , Osteoporosis Posmenopáusica/complicaciones , Ligando RANK/efectos adversos , Ligando RANK/farmacología
20.
Praxis (Bern 1994) ; 100(14): 821-32, 2011 Jul 06.
Artículo en Alemán | MEDLINE | ID: mdl-21732293

Asunto(s)
Osteoporosis Posmenopáusica/diagnóstico , Absorciometría de Fotón , Anciano , Algoritmos , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Conservadores de la Densidad Ósea/efectos adversos , Conservadores de la Densidad Ósea/uso terapéutico , Calcitonina/efectos adversos , Calcitonina/uso terapéutico , Calcio/administración & dosificación , Denosumab , Difosfonatos/efectos adversos , Difosfonatos/uso terapéutico , Femenino , Fracturas del Cuello Femoral/diagnóstico , Fracturas del Cuello Femoral/etiología , Fracturas del Cuello Femoral/prevención & control , Humanos , Masculino , Compuestos Organometálicos/efectos adversos , Compuestos Organometálicos/uso terapéutico , Osteoporosis Posmenopáusica/tratamiento farmacológico , Osteoporosis Posmenopáusica/etiología , Fracturas Osteoporóticas/diagnóstico , Fracturas Osteoporóticas/etiología , Fracturas Osteoporóticas/prevención & control , Ligando RANK/efectos adversos , Ligando RANK/uso terapéutico , Factores de Riesgo , Moduladores Selectivos de los Receptores de Estrógeno/efectos adversos , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Enfermedades de la Columna Vertebral/diagnóstico , Enfermedades de la Columna Vertebral/etiología , Enfermedades de la Columna Vertebral/prevención & control , Teriparatido/efectos adversos , Teriparatido/uso terapéutico , Tiofenos/efectos adversos , Tiofenos/uso terapéutico , Vitamina D/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...