Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Regul Toxicol Pharmacol ; 81: 212-222, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27569204

RESUMEN

Romosozumab is a humanized immunoglobulin G2 monoclonal antibody that binds and blocks the action of sclerostin, a protein secreted by the osteocyte and an extracellular inhibitor of canonical Wnt signaling. Blockade of sclerostin binding to low-density lipoprotein receptor-related proteins 5 and 6 (LRP5 and LRP6) allows Wnt ligands to activate canonical Wnt signaling in bone, increasing bone formation and decreasing bone resorption, making sclerostin an attractive target for osteoporosis therapy. Because romosozumab is a bone-forming agent and an activator of canonical Wnt signaling, questions have arisen regarding a potential carcinogenic risk. Weight-of-evidence factors used in the assessment of human carcinogenic risk of romosozumab included features of canonical Wnt signaling, expression pattern of sclerostin, phenotype of loss-of-function mutations in humans and mice, mode and mechanism of action of romosozumab, and findings from romosozumab chronic toxicity studies in rats and monkeys. Although the weight-of-evidence factors supported that romosozumab would pose a low carcinogenic risk to humans, the carcinogenic potential of romosozumab was assessed in a rat lifetime study. There were no romosozumab-related effects on tumor incidence in rats. The findings of the lifetime study and the weight-of-evidence factors collectively indicate that romosozumab administration would not pose a carcinogenic risk to humans.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Neoplasias/inducido químicamente , Animales , Anticuerpos Monoclonales/administración & dosificación , Pruebas de Carcinogenicidad , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratas , Medición de Riesgo
2.
Calcif Tissue Int ; 96(1): 45-55, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25467010

RESUMEN

The purpose of this study is to estimate the efficacy of eldecalcitol (1α, 25-Dihydroxy-2ß- (3-hydroxypropyloxy) vitamin D3; ELD) on bone metabolism after long-term administration. Six-month-old Wistar-Imamichi rats were ovariectomized (OVX) and administered ELD orally at doses of 7.5, 15, or 30 ng/kg daily. Bone mineral density (BMD), urinary excretion of deoxypyridinoline (DPD), a bone resorption marker, and serum total alkaline phosphatase (ALP), a surrogate marker of bone formation, were assessed after 3, 6, and 12 months of treatment. After 12 months of treatment, the biomechanical strength of the L4 lumbar vertebra and femoral shaft was measured, and bone histomorphometry was performed on the L3 lumbar vertebra and the tibia diaphysis. ELD prevented OVX-induced decreases in BMD of the lumbar vertebrae and femur throughout the treatment period. ELD significantly suppressed OVX-induced increases in urinary DPD excretion throughout the treatment period with minimal effects on ALP. OVX resulted in significant decreases in ultimate load and stiffness of the L4 lumbar vertebra and femoral shaft, and ELD significantly prevented the reduction in these biomechanical parameters. Bone histomorphometry at the L3 lumbar vertebra revealed that OVX induced increases in bone resorption parameters (osteoclast surface and osteoclast number) and bone formation parameters (osteoblast surface, osteoid surface, and bone formation rate), and ELD suppressed these parameters after 12 months treatment. Activation frequency, which was elevated in the OVX/vehicle group, was significantly suppressed to baseline levels in ELD-treated groups, indicating that ELD maintained bone turnover at a normal level. ELD also prevented OVX-induced deterioration of microstructure in trabecular and cortical bone. These results indicated that long-term treatment of OVX rats with ELD suppressed bone turnover, and prevented OVX-induced bone loss, deterioration of bone microstructure, and reduction in bone biomechanical strength.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Huesos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Vitamina D/análogos & derivados , Animales , Densidad Ósea/fisiología , Resorción Ósea/tratamiento farmacológico , Femenino , Fémur/efectos de los fármacos , Vértebras Lumbares/efectos de los fármacos , Ovariectomía/métodos , Ratas , Tiempo , Vitamina D/administración & dosificación , Vitamina D/uso terapéutico
3.
J Bone Miner Metab ; 33(6): 625-41, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25534548

RESUMEN

Pioglitazone, the peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonist is an effective therapy for type 2 diabetes, but has been associated with increased risk for bone fracture. Preclinical studies suggest that PPAR-α agonists (e.g., fenofibrate) increase bone mineral density/content, although clinical data on bone effects of fibrates are lacking. We investigated the effects of pioglitazone (10 mg/kg/day) and fenofibrate (25 mg/kg/day) on bone strength and bone histomorphometric parameters in osteopenic ovariectomized (OVX) rats. An additional group of rats received a combination of pioglitazone + fenofibrate to mimic the effects of a dual PPAR-α/γ agonist. The study consisted of a 13-week treatment phase followed by a 6-week treatment-free recovery period. Pioglitazone significantly reduced biomechanical strength at the lumbar spine and femoral neck compared with rats administered fenofibrate. Co-treatment with pioglitazone + fenofibrate had no significant effect on bone strength in comparison with OVX vehicle controls. Histomorphometric analysis of the proximal tibia revealed that pioglitazone suppressed bone formation and increased bone resorption at both cancellous and cortical bone sites relative to OVX vehicle controls. In contrast, fenofibrate did not affect bone resorption and only slightly suppressed bone formation. Discontinuation of pioglitazone treatment, both in the monotherapy and in the combination therapy arms, resulted in restoration of bone formation and resorption rates, demonstrating reversibility of effects. The above data support the concept that dual activation of PPAR-γ and PPAR-α attenuates the negative effects of PPAR-γ agonism on bone strength.


Asunto(s)
Huesos/patología , Huesos/fisiopatología , Fenofibrato/administración & dosificación , Fenofibrato/farmacología , Ovariectomía , Tiazolidinedionas/administración & dosificación , Tiazolidinedionas/farmacología , Absorciometría de Fotón , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Fuerza Compresiva/efectos de los fármacos , Densitometría , Diáfisis/diagnóstico por imagen , Diáfisis/efectos de los fármacos , Diáfisis/patología , Diáfisis/fisiopatología , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/patología , Fémur/fisiopatología , Cuello Femoral/diagnóstico por imagen , Cuello Femoral/efectos de los fármacos , Cuello Femoral/patología , Cuello Femoral/fisiopatología , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Pioglitazona , Ratas Sprague-Dawley , Tibia/diagnóstico por imagen , Tibia/efectos de los fármacos , Tibia/patología , Tibia/fisiopatología , Tomografía Computarizada por Rayos X
4.
Birth Defects Res B Dev Reprod Toxicol ; 98(5): 374-90, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24323897

RESUMEN

BACKGROUND: The timing and duration of letrozole administration was designed to encompass the majority of postnatal development in the rat with the intent of evaluating the potential for a broad range of effects but with emphasis on expected effects on skeletal maturation. METHODS: Sprague-Dawley rats were administered letrozole via oral gavage at doses of 0.003, 0.03, and 0.3 mg/kg/day beginning on postpartum day (PPD) 7 through 91 followed by a 6-week recovery period. Clinical signs, body weight, food consumption, developmental endpoints, bone, ophthalmology, behavioral assessments, clinical/anatomic pathology, toxicokinetics, and reproductive assessments were conducted. RESULTS: Growth (body weight gain and crown-to-rump length) and food consumption were increased in females at ≥0.03 mg/kg/day and decreased in males at ≥0.003 mg/kg/day. Delayed sexual maturation in both sexes and adverse effects on reproductive function occurred at all doses. Effects on bone growth and maturation were noted in both sexes at all doses. Evidence of recovery was noted for males at 0.003 mg/kg/day and females at 0.003 and 0.03 mg/kg/day upon withdrawal of treatment. Histopathological changes in the pituitary-adrenal-gonadal axis correlated with effects on reproductive function. CONCLUSIONS: The observed effects in juvenile rats were considered predictable and primarily related to the mechanism of action of letrozole upon estrogen synthesis.


Asunto(s)
Inhibidores de la Aromatasa/toxicidad , Nitrilos/toxicidad , Triazoles/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Desarrollo Óseo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Letrozol , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos , Espermatozoides/efectos de los fármacos
5.
Microbiome ; 10(1): 105, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35799219

RESUMEN

BACKGROUND: Inflammatory bowel diseases (IBDs) including Crohn's disease (CD) and ulcerative colitis (UC) are characterized by chronic and debilitating gut inflammation. Altered bacterial communities of the intestine are strongly associated with IBD initiation and progression. The gut virome, which is primarily composed of bacterial viruses (bacteriophages, phages), is thought to be an important factor regulating and shaping microbial communities in the gut. While alterations in the gut virome have been observed in IBD patients, the contribution of these viruses to alterations in the bacterial community and heightened inflammatory responses associated with IBD patients remains largely unknown. RESULTS: Here, we performed in vivo microbial cross-infection experiments to follow the effects of fecal virus-like particles (VLPs) isolated from UC patients and healthy controls on bacterial diversity and severity of experimental colitis in human microbiota-associated (HMA) mice. Shotgun metagenomics confirmed that several phages were transferred to HMA mice, resulting in treatment-specific alterations in the gut virome. VLPs from healthy and UC patients also shifted gut bacterial diversity of these mice, an effect that was amplified during experimental colitis. VLPs isolated from UC patients specifically altered the relative abundance of several bacterial taxa previously implicated in IBD progression. Additionally, UC VLP administration heightened colitis severity in HMA mice, as indicated by shortened colon length and increased pro-inflammatory cytokine production. Importantly, this effect was dependent on intact VLPs. CONCLUSIONS: Our findings build on recent literature indicating that phages are dynamic regulators of bacterial communities in the gut and implicate the intestinal virome in modulating intestinal inflammation and disease. Video Abstract.


Asunto(s)
Bacteriófagos , Colitis Ulcerosa , Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Animales , Bacterias/genética , Bacteriófagos/genética , Colitis/terapia , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/terapia , Inflamación , Enfermedades Inflamatorias del Intestino/microbiología , Ratones
6.
MAbs ; 14(1): 1979447, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34923919

RESUMEN

Targeting immune checkpoint receptors expressed in the T cell synapse induces active and long-lasting antitumor immunity in preclinical tumor models and oncology patients. However, traditional nonhuman primate (NHP) studies in healthy animals have thus far demonstrated little to no pharmacological activity or toxicity for checkpoint inhibitors (CPIs), likely due to a quiescent immune system. We developed a NHP vaccine challenge model in Mauritius cynomolgus monkey (MCMs) that elicits a strong CD8+ T cell response to assess both pharmacology and safety within the same animal. MHC I-genotyped MCMs were immunized with three replication incompetent adenovirus serotype 5 (Adv5) encoding Gag, Nef and Pol simian immunodeficiency virus (SIV) proteins administered 4 weeks apart. Immunized animals received the anti-PD-L1 atezolizumab or an immune checkpoint-targeting bispecific antibody (mAbX) in early development. After a single immunization, Adv5-SIVs induced T-cell activation as assessed by the expression of several co-stimulatory and co-inhibitory molecules, proliferation, and antigen-specific T-cell response as measured by a Nef-dependent interferon-γ ELIspot and tetramer analysis. Administration of atezolizumab increased the number of Ki67+ CD8+ T cells, CD8+ T cells co-expressing TIM3 and LAG3 and the number of CD4+ T cells co-expressing 4-1BB, BTLA, and TIM3 two weeks after vaccination. Both atezolizumab and mAbX extended the cytolytic activity of the SIV antigen-specific CD8+ T cell up to 8 weeks. Taken together, this vaccine challenge model allowed the combined study of pharmacology and safety parameters for a new immunomodulatory protein-based therapeutic targeting CD8+ T cells in an NHP model.


Asunto(s)
Adenoviridae , Linfocitos T CD8-positivos/inmunología , Vacunas contra el SIDAS , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Evaluación de Medicamentos , Macaca fascicularis , Masculino , Vacunas contra el SIDAS/genética , Vacunas contra el SIDAS/inmunología , Vacunas contra el SIDAS/farmacología , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/genética
7.
J Bone Miner Res ; 22(1): 55-63, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17014384

RESUMEN

UNLABELLED: We examined the effects of 60 days of co-treatment of PTH with either OPG or alendronate in oophorectomized mice. Compared with PTH alone, co-treatment of PTH with either of these two mechanistically distinct anti-catabolics improved bone volume, mechanical strength, and appendicular and axial mineralization and prolonged the beneficial effect of PTH on BMD. INTRODUCTION: Conflicting evidence exists as to whether the anabolic effect of PTH is inhibited by the action of anti-catabolics. To examine this issue, we assessed the effects of alendronate and osteoprotegerin (OPG), two anti-catabolics with different modes of action, on the anabolic activity of PTH(1-34) in the skeleton of 4-month-old oophorectomized mice. MATERIALS AND METHODS: Mice treated with vehicle alone (PBS), alendronate alone (100 microg/kg/week), OPG alone (10 mg/kg twice a week), or PTH alone (80 microg/kg/day) were compared with each other and with animals administered PTH plus alendronate or PTH plus OPG. We assessed lumbar spine and femoral BMD at 0, 30, and 60 days. Contact radiography, histology, and histomorphometry, three-point bending assay of the femur, and serum osteocalcin and TRACP5b assays were performed at 2 months. RESULTS: Although alendronate and OPG each suppressed bone turnover, at the doses used, this was more profound with OPG. Increases in lumbar spine and femoral BMD and in trabecular bone volume were at least as great with OPG as with alendronate, and mechanical indices of femoral bone strength improved only with OPG. Both produced a plateau in spine and femoral BMD increases by 30 days. Co-treatment of PTH with each anti-catabolic produced additive increases in BMD in the femur and supra-additive increases in the lumbar spine with no plateau effects. Neither anti-catabolic impeded the PTH-induced increase in bone volume or the increase in mechanical strength of the femur. CONCLUSIONS: These studies show that the highly potent anti-catabolic OPG can produce dramatic increases in BMD and bone strength; that the temporal pattern of activity of bone formation and resorption modulators may have major influence on net skeletal accrual; and that, depending on timing, inhibition of osteoclastic activity may markedly augment the anabolic action of PTH.


Asunto(s)
Alendronato/farmacología , Huesos/fisiología , Osteoprotegerina/farmacología , Hormona Paratiroidea/farmacología , Columna Vertebral/fisiología , Animales , Huesos/efectos de los fármacos , Femenino , Fémur/efectos de los fármacos , Fémur/fisiología , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Columna Vertebral/efectos de los fármacos
8.
Endocrinology ; 148(6): 2778-87, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17379647

RESUMEN

Previous studies have indicated that bisphosphonate pretreatment can inhibit the anabolic actions of PTH. We examined the capacity of two anticatabolic agents with different mechanisms of action, alendronate and osteoprotegerin (OPG), to influence the anabolic activity of PTH. Oophorectomized mice were pretreated for 30 d with alendronate or OPG and then treated for 30 d with the respective anticatabolic alone or the respective anticatabolic plus PTH(1-34). Bones were analyzed by bone mineral density (BMD), microcomputed tomography, histology and histomorphometry, and biochemical bone markers. OPG pretreatment produced a greater inhibition of bone turnover and a greater increase in bone than alendronate. Increases in bone were sustained during subsequent treatment with vehicle or continued administration of the anticatabolic. Pretreatment with each anticatabolic blunted the capacity of PTH to increase BMD and bone volume and continued treatment with each anticatabolic agent also reduced the effectiveness of PTH. Although both anticatabolics decreased the maximal PTH effect, BMD and bone volume increased more when PTH was added than when only anticatabolics were used. These results demonstrate that mechanistically distinct anticatabolics may reduce PTH efficacy, that the characteristics of this inhibition may reflect the different modes of action of the anticatabolics, but that the addition of PTH still provides a skeletal benefit even if the anabolic effect is submaximal.


Asunto(s)
Alendronato/uso terapéutico , Resorción Ósea/prevención & control , Huesos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Alendronato/administración & dosificación , Animales , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/uso terapéutico , Huesos/metabolismo , Quimioterapia Combinada , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteoprotegerina/administración & dosificación , Osteoprotegerina/uso terapéutico , Ovariectomía
9.
Endocrinology ; 148(6): 2613-21, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17332067

RESUMEN

Osteoporosis is a leading public health problem. Although a major cause in women is thought to be a decline in estrogen, it has recently been proposed that FSH or follitropin is required for osteoporotic bone loss. We examined the FSH receptor null mouse (FORKO mouse) to determine whether altered ovarian function could induce bone loss independent of FSH action. By 3 months of age, FORKO mice developed age-dependent declines in bone mineral density and trabecular bone volume of the lumbar spine and femur, which could be partly reversed by ovarian transplantation. Bilateral ovariectomy reduced elevated circulating testosterone levels in FORKO mice and decreased bone mass to levels indistinguishable from those in ovariectomized wild-type controls. Androgen receptor blockade and especially aromatase inhibition each produced bone volume reductions in the FORKO mouse. The results indicate that ovarian secretory products, notably estrogen, and peripheral conversion of ovarian androgen to estrogen can alter bone homeostasis independent of any bone resorptive action of FSH.


Asunto(s)
Huesos/fisiología , Hormona Folículo Estimulante/fisiología , Homeostasis , Ovario/fisiología , Andrógenos/farmacología , Animales , Densidad Ósea/genética , Huesos/efectos de los fármacos , Huesos/metabolismo , Femenino , Homeostasis/genética , Masculino , Ratones , Ratones Noqueados , Osteoporosis/patología , Ovariectomía , Ovario/fisiopatología , Ovario/trasplante , Receptores de HFE/genética
10.
J Orthop Res ; 34(1): 72-80, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26178170

RESUMEN

The effects of the cathepsin K inhibitor odanacatib (ODN) on fracture healing were monitored for ~6 and 15 weeks post-fracture in two separate studies using the unilateral transverse mid-ulnar osteotomy model in skeletally mature female rabbits. Rabbits were pre-treated for 3-4 weeks with vehicle (Veh), ODN (2 mg/kg, po, daily), or alendronate (ALN) (0.3 mg/kg, sc, twice-weekly) prior to osteotomy. In Study 1, the animals were maintained on the same respective treatment for ~6 weeks. In Study 2, the animals were also continued on the same therapy or switched from Veh to ODN or ODN to Veh for 15 weeks. No treatment-related impairment of fracture union was seen by qualitative histological assessments in the first study. Cartilage retention was detected in the calluses of ALN-treated rabbits at week-6, while calluses in the ODN and Veh groups contained bony tissue with significantly less residual cartilage. ODN treatment also markedly increased the number of cathepsin K-(+) osteoclasts in the callus, indicating enhanced callus remodeling. From the second study, ex vivo DXA and pQCT confirmed that ODN treatment pre- and post-osteotomy increased callus bone mineral content and bone mineral density (BMD) versus Veh (p < 0.001) and discontinuation of ODN post-surgery returned callus BMD to Veh. Peak load of ODN- or ALN-treated calluses were comparable to Veh. ODN increased callus yield load (20%, p = 0.056) and stiffness (26%, p < 0.05) versus Veh. These studies demonstrated that ODN increased mineralized callus during the early phase of fracture repair without impairing callus formation or biomechanical integrity at the fracture site.


Asunto(s)
Compuestos de Bifenilo/uso terapéutico , Callo Óseo/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Curación de Fractura/efectos de los fármacos , Alendronato/farmacología , Alendronato/uso terapéutico , Animales , Compuestos de Bifenilo/farmacología , Femenino , Osteotomía , Conejos , Distribución Aleatoria , Cúbito
11.
J Bone Miner Res ; 20(4): 672-81, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15765187

RESUMEN

UNLABELLED: Human osteoblasts produce PGD(2), which acts on the DP receptor to decrease osteoprotegerin production and on the CRTH2 receptor to decrease RANKL expression and to induce osteoblast chemotaxis. These results indicate that activation of CRTH2 may lead to an anabolic response in bone. INTRODUCTION: Whereas the actions of prostaglandin (PG)E(2) as a modulator of bone and osteoblast function are relatively well characterized, little is known about PGD(2) and bone metabolism. The objectives of this study were to determine if human osteoblasts can produce PGD(2), which prostaglandin D(2) synthases are implicated in this synthesis, to identify the PGD(2) receptors (DP and CRTH2) on these cells and to characterize the biological effects resulting from their activation. MATERIALS AND METHODS: RT-PCR analysis and immunohistochemistry were used to detect PGD(2) receptor and synthases in cultured human osteoblasts. Immunohistochemistry was used to identify the synthases and receptors in human bone tissue. Intracellular cAMP and calcium levels were determined to verify receptor activation. The cells were stimulated with PGD(2) or the specific agonists BW 245C (DP) and DK-PGD(2) (CRTH2), and the resulting effects on osteoprotegerin (OPG) secretion, RANKL expression, and chemotaxis were determined. Osteoblast production of PGD(2) was evaluated by measuring PGD(2) in the culture supernatants after stimulation with interleukin (IL)-1, TNF-alpha, PTH, vascular endothelial growth factor (VEGF), and insulin-like growth factor I (IGF-I). RESULTS: Human osteoblasts in culture generated PGD(2) when stimulated. Both osteoblasts in culture and in situ present the lipocalin-type PGD(2) synthase only. Both DP and CRTH2 receptors were present in human osteoblasts in culture and in situ. Stimulation of DP resulted in an increase in cAMP, whereas CRTH2 increased the intracellular calcium level. OPG production was reduced by 60% after DP receptor stimulation, whereas CRTH2 receptor stimulation decreased RANKL expression on human osteoblasts. As reported for other cell types, CRTH2 was a potent inducer of chemotaxis for human osteoblasts in culture. CONCLUSIONS: Human osteoblasts in culture produce PGD(2) under biologically relevant stimuli through the lipocalin-type PGD(2) synthase (L-PGDS) pathway. As an autacoid, PGD(2) can act on DP and CRTH2 receptors, both present on these cells. Specific activation of CRTH2 could lead directly and indirectly to an anabolic response in bone.


Asunto(s)
Quimiotaxis , Osteoblastos/metabolismo , Prostaglandina D2/biosíntesis , Receptores Inmunológicos/fisiología , Receptores de Prostaglandina/fisiología , Huesos/metabolismo , Proteínas Portadoras/metabolismo , AMP Cíclico/metabolismo , Glicoproteínas/metabolismo , Sustancias de Crecimiento/farmacología , Humanos , Hidantoínas/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Interleucina-1/farmacología , Glicoproteínas de Membrana/metabolismo , Osteoblastos/efectos de los fármacos , Osteoprotegerina , Prostaglandina D2/genética , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Prostaglandina/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
12.
J Bone Miner Res ; 30(4): 657-69, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25369992

RESUMEN

Postmenopausal osteoporosis is a chronic disease wherein increased bone remodeling reduces bone mass and bone strength. Antiresorptive agents including bisphosphonates are commonly used to mitigate bone loss and fracture risk. Osteoclast inhibition via denosumab (DMAb), a RANKL inhibitor, is a newer approach for reducing fracture risk in patients at increased risk for fracture. The safety of transitioning from bisphosphonate therapy (alendronate; ALN) to DMAb was examined in mature ovariectomized (OVX) cynomolgus monkeys (cynos). One day after OVX, cynos (7-10/group) were treated with vehicle (VEH, s.c.), ALN (50 µg/kg, i.v., twice monthly) or DMAb (25 mg/kg/month, s.c.) for 12 months. Other animals received VEH or ALN for 6 months and then transitioned to 6 months of DMAb. DMAb caused significantly greater reductions in serum CTx than ALN, and transition from ALN to DMAb caused further reductions relative to continued ALN. DMAb and ALN decreased serum calcium (Ca), and transition from ALN to DMAb resulted in a lesser decline in Ca relative to DMAb or to VEH-DMAb transition. Bone histomorphometry indicated significantly reduced trabecular and cortical remodeling with DMAb or ALN. Compared with ALN, DMAb caused greater reductions in osteoclast surface, eroded surface, cortical porosity and fluorochrome labeling, and transition from ALN to DMAb reduced these parameters relative to continued ALN. Bone mineral density increased in all active treatment groups relative to VEH controls. Destructive biomechanical testing revealed significantly greater vertebral strength in all three groups receiving DMAb, including those receiving DMAb after ALN, relative to VEH controls. Bone mass and strength remained highly correlated in all groups at all tested skeletal sites, consistent with normal bone quality. These data indicate that cynos transitioned from ALN to DMAb exhibited reduced bone resorption and cortical porosity, and increased BMD and bone strength, without deleterious effects on Ca homeostasis or bone quality.


Asunto(s)
Alendronato/farmacología , Conservadores de la Densidad Ósea/farmacología , Huesos/efectos de los fármacos , Calcio/metabolismo , Denosumab/farmacología , Homeostasis/efectos de los fármacos , Ovariectomía , Absorciometría de Fotón , Alendronato/efectos adversos , Animales , Conservadores de la Densidad Ósea/efectos adversos , Huesos/fisiología , Denosumab/efectos adversos , Femenino , Macaca fascicularis
13.
Bone ; 66: 72-81, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24928497

RESUMEN

Cathepsin K (CatK) is a cysteine protease, expressed predominantly in osteoclasts (OC) which degrades demineralized bone matrix. Novel selective inhibitors of CatK are currently being developed for the treatment of postmenopausal osteoporosis. Pharmacological inhibition of CatK reduces OC resorption activity while preserving bone formation in preclinical models. Disruption of the CatK gene in mice also results in high bone mass due to impaired bone resorption and elevated formation. Here, we assessed mid-shaft femoral fracture healing in 8-10week old CatK knock-out (KO) versus wild type (WT) mice. Fracture healing and callus formation were determined in vivo weekly via X-ray, and ex vivo at days 14, 18, 28 and 42 post-fracture by radiographic scoring, micro-computed tomography (µCT), histomorphometry and terminal mechanical four point bend strength testing. Radiological evaluation indicated accelerated bone healing and remodeling for CatK KO animals based on increased total radiographic scores that included callus opacity and bridging at days 28 and 42 post-fracture. Micro-CT based total callus volume was similar in CatK KO and WT mice at day 14. Callus size in CatK KO mice was 25% smaller than that in WT mice at day 18, statistically significant by day 28 and exhibited significantly higher mineralized tissue volume and volumetric BMD as compared to WT by day 18 onward. Osteoclast surface and osteoid surface trended higher in CatK KO calluses at all time-points and osteoblast number was also significantly increased at day 28. Increased CatK KO callus mineral density was reflected in significant increases in peak load and stiffness over WT at day 42 post-fracture. Regression analysis indicated a positive correlation (r=0.8671; p<0.001) between callus BMC and peak load indicating normal mineral properties in CatK KO calluses. Taken together, gene deletion of cathepsin K in mice accelerated callus size resolution, significantly increased callus mineralized mass, and improved mechanical strength as compared to wild type mice.


Asunto(s)
Callo Óseo/patología , Callo Óseo/fisiopatología , Calcificación Fisiológica , Catepsina K/deficiencia , Fracturas del Fémur/patología , Fracturas del Fémur/fisiopatología , Animales , Fenómenos Biomecánicos , Remodelación Ósea , Catepsina K/metabolismo , Recuento de Células , Femenino , Fracturas del Fémur/diagnóstico por imagen , Curación de Fractura , Eliminación de Gen , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Osteoblastos/patología , Osteoclastos/patología , Radiografía
14.
J Bone Miner Res ; 29(8): 1847-58, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24591096

RESUMEN

Treatment with the cathepsin K (CatK) inhibitor odanacatib (ODN) protects against bone loss and maintains normal biomechanical properties in the spine and hip of ovariectomized (OVX) preclinical models. Here, we characterized the effects of ODN on the dynamics of cortical modeling and remodeling, and dimension and strength of the central femur in adult OVX-rhesus monkeys. Animals were treated with vehicle or ODN (6 or 30 mg/kg, once per day [q.d., p.o.]) in prevention mode for 21 months. Calcein and tetracycline double-labeling were given at 12 and 21 months, and the femoral cross-sections were subjected to dynamic histomorphometric and cement line analyses. ODN treatment significantly increased periosteal and endocortical bone formation (BFR/BS), accompanied with an increase in endocortical mineralizing surface (102%, p < 0.01) with the 6 mg/kg dose. ODN at both doses reduced remodeling hemiosteon numbers by 51% and 66% (p < 0.05), respectively, and ODN 30 mg/kg numerically reduced activation frequency without affecting wall thickness. On the same endocortical surface, ODN increased all modeling-based parameters, while reducing intracortical remodeling, consistent with the observed no treatment effects on cortical porosity. ODN 30 mg/kg markedly increased cortical thickness (CtTh, p < 0.001) and reduced marrow area (p < 0.01). Lastly, ODN treatment increased femoral structural strength (p < 0.001). Peak load was positively correlated with the increases in bone mineral content (BMC) (r(2) = 0.9057, p < 0.0001) and CtTh (r2 = 0.6866, p < 0.0001). Taken together, by reducing cortical remodeling-based and stimulating modeling-based bone formation, ODN significantly improved cortical dimension and strength in OVX monkeys. This novel mechanism of CatK inhibition in stimulating cortical formation suggests that ODN represents a novel therapeutic approach for the treatment of osteoporosis.


Asunto(s)
Compuestos de Bifenilo/farmacología , Densidad Ósea/efectos de los fármacos , Remodelación Ósea/efectos de los fármacos , Catepsina K/antagonistas & inhibidores , Osteogénesis/efectos de los fármacos , Animales , Compuestos de Bifenilo/administración & dosificación , Femenino , Cadera/patología , Macaca mulatta , Ovariectomía , Columna Vertebral/efectos de los fármacos
15.
J Bone Miner Res ; 28(7): 1653-65, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23456892

RESUMEN

Rosiglitazone (RSG) is an antidiabetic drug that has been associated with increased peripheral fractures, primarily in postmenopausal women. In this report, we investigated the underlying mechanisms of RSG-associated bone loss in ovariectomized (OVX) rats and determined whether changes in bone parameters associated with RSG administration are reversible on treatment cessation or preventable by coadministration with an antiresorptive agent. Nine-month-old Sprague-Dawley rats underwent OVX or sham operation. Sham-operated rats received oral vehicle only; OVX animals were randomized to receive vehicle, RSG, alendronate (ALN), or RSG plus ALN for 12 weeks. All treatment started the day after ovariectomy. After the 12-week treatment period, the OVX and RSG groups also underwent an 8-week treatment-free recovery period. Bone densitometry measurements, bone turnover markers, biomechanical testing, and histomorphometric analysis were conducted. Microcomputed tomography was also used to investigate changes in microarchitecture. RSG significantly increased deoxypyridinoline levels compared with OVX. Significant exacerbation of OVX-induced loss of bone mass, strength, and microarchitectural deterioration was observed in RSG-treated OVX animals compared with OVX controls. These effects were observed predominantly at sites rich in trabecular bone, with less pronounced effects in cortical bone. Coadministration of RSG and ALN prevented the bone loss associated with RSG treatment. Following cessation of RSG treatment, effects on bone mass and strength showed evidence of reversal. Thus, treatment of OVX rats with RSG results in loss of bone mass and strength, primarily at sites rich in trabecular bone, mainly due to increased bone resorption. These effects can be prevented by concomitant treatment with ALN and may be reversed following discontinuation of RSG.


Asunto(s)
Alendronato/farmacología , Conservadores de la Densidad Ósea/farmacología , Densidad Ósea/efectos de los fármacos , Hipoglucemiantes/farmacología , Osteoporosis Posmenopáusica/tratamiento farmacológico , Tiazolidinedionas/farmacología , Animales , Femenino , Fracturas Óseas/etiología , Fracturas Óseas/metabolismo , Humanos , Osteoporosis Posmenopáusica/complicaciones , Osteoporosis Posmenopáusica/metabolismo , Ovariectomía , Ratas , Ratas Sprague-Dawley , Rosiglitazona , Factores de Tiempo , Microtomografía por Rayos X
17.
J Endocrinol ; 212(2): 179-86, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22062085

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) γ agonists, such as pioglitazone (Pio), improve glycemia and lipid profile but are associated with bone loss and fracture risk. Data regarding bone effects of PPARα agonists (including fenofibrate (Feno)) are limited, although animal studies suggest that Feno may increase bone mass. This study investigated the effects of a 13-week oral combination treatment with Pio (10 mg/kg per day)+Feno (25 mg/kg per day) on body composition and bone mass parameters compared with Pio or Feno alone in adult ovariectomized (OVX) rats, with a 4-week bone depletion period, followed by a 6-week treatment-free period. Treatment of OVX rats with Pio+Feno resulted in ∼50% lower fat mass gain compared with Pio treatment alone. Combination treatment with Pio+Feno partially prevented Pio-induced loss of bone mineral content (∼45%) and bone mineral density (BMD; ∼60%) at the lumbar spine. Similar effects of treatments were observed at the femur, most notably at sites rich in trabecular bone. At the proximal tibial metaphysis, concomitant treatment with Pio+Feno prevented Pio exacerbation of ovariectomy-induced loss of trabecular bone, resulting in BMD values in the Pio+Feno group comparable to OVX controls. Discontinuation of Pio or Feno treatment of OVX rats was associated with partial reversal of effects on bone loss or bone mass gain, respectively, while values in the Pio+Feno group remained comparable to OVX controls. These data suggest that concurrent/dual agonism of PPARγ and PPARα may reduce the negative effects of PPARγ agonism on bone mass.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/prevención & control , Fenofibrato/uso terapéutico , Osteoporosis Posmenopáusica/tratamiento farmacológico , PPAR alfa/agonistas , PPAR gamma/agonistas , Tiazolidinedionas/efectos adversos , Adiposidad/efectos de los fármacos , Animales , Biomarcadores/sangre , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/administración & dosificación , Resorción Ósea/inducido químicamente , Resorción Ósea/etiología , Huesos/química , Huesos/efectos de los fármacos , Colágeno Tipo I/sangre , Quimioterapia Combinada , Femenino , Fenofibrato/administración & dosificación , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/uso terapéutico , Hipolipemiantes/administración & dosificación , Hipolipemiantes/efectos adversos , Hipolipemiantes/uso terapéutico , Osteocalcina/sangre , Osteoporosis Posmenopáusica/sangre , Osteoporosis Posmenopáusica/fisiopatología , Ovariectomía , Péptidos/sangre , Pioglitazona , Distribución Aleatoria , Ratas , Tiazolidinedionas/administración & dosificación , Tiazolidinedionas/uso terapéutico
18.
Bone ; 49(3): 376-86, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21658483

RESUMEN

A novel approach to menopausal therapy is the tissue selective estrogen complex (TSEC) that partners bazedoxifene (BZA) with conjugated estrogens (CE). We examined the effects of daily treatment with BZA 0.3mg/kg, CE 2.5mg/kg, or combined BZA/CE (BZA 0.1, 0.3, or 1.0mg/kg with CE 2.5mg/kg) over 12months on bone mass, bone architecture and strength, and biochemical markers of bone turnover in ovariectomized (OVX) female Sprague-Dawley rats vs OVX control rats. Total cholesterol and uterine weights were also evaluated. All BZA/CE dose combinations prevented ovariectomy-induced increases in bone turnover and significantly increased bone mineral density (BMD) at the lumbar spine, proximal femur, and tibia compared with OVX controls. All BZA/CE doses evaluated also prevented many of the ovariectomy-induced changes of the static and dynamic parameters of the cortical compartment of the tibia and the cancellous compartment of the L1 and L2 vertebrae. All BZA/CE doses increased biomechanical strength at the lumbar spine (L4) compared with OVX animals. The co-administration of BZA 0.3 and 1.0mg/kg/day with CE 2.5mg/kg/day showed a dose-dependent reduction in uterine wet weight compared with administration of CE alone. All BZA/CE doses significantly lowered total cholesterol levels compared with OVX controls. In conclusion, 12months of treatment with BZA/CE in OVX rats effectively maintained BMD, bone microstructure, and bone quality; and the pairing of BZA with CE prevented CE-induced uterine stimulation.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Huesos/efectos de los fármacos , Huesos/fisiología , Estrógenos Conjugados (USP)/farmacología , Estrógenos/farmacología , Indoles/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Biomarcadores/metabolismo , Densidad Ósea/efectos de los fármacos , Huesos/anatomía & histología , Femenino , Fémur/anatomía & histología , Fémur/efectos de los fármacos , Fémur/fisiología , Humanos , Vértebras Lumbares/anatomía & histología , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/fisiología , Tamaño de los Órganos , Ovariectomía , Ratas , Ratas Sprague-Dawley , Tibia/anatomía & histología , Tibia/efectos de los fármacos , Tibia/fisiología , Útero/anatomía & histología , Útero/efectos de los fármacos
19.
J Bone Miner Res ; 26(2): 252-62, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20734451

RESUMEN

Two cathepsin K inhibitors (CatKIs) were compared with alendronate (ALN) for their effects on bone resorption and formation in ovariectomized (OVX) rabbits. The OVX model was validated by demonstrating significant loss (9.8% to 12.8%) in lumbar vertebral bone mineral density (LV BMD) in rabbits at 13-weeks after surgery, which was prevented by estrogen or ALN. A potent CatKI, L-006235 (L-235), dosed at 10 mg/kg per day for 27 weeks, significantly decreased LV BMD loss (p < .01) versus OVX-vehicle control. ALN reduced spine cancellous mineralizing surface by 70%, whereas L-235 had no effect. Similarly, endocortical bone-formation rate and the number of double-labeled Haversian canals in the femoral diaphysis were not affected by L-235. To confirm the sparing effects of CatKI on bone formation, odanacatib (ODN) was dosed in food to achieve steady-state exposures of 4 or 9 µM/day in OVX rabbits for 27 weeks. ODN at both doses prevented LV BMD loss (p < .05 and p < .001, respectively) versus OVX-vehicle control to levels comparable with sham or ALN. ODN also dose-dependently increased BMD at the proximal femur, femoral neck, and trochanter. Similar to L-235, ODN did not reduce bone formation at any bone sites studied. The positive and highly correlative relationship of peak load to bone mineral content in the central femur and spine suggested that ODN treatment preserved normal biomechanical properties of relevant skeletal sites. Although CatKIs had similar efficacy to ALN in preventing bone loss in adult OVX rabbits, this novel class of antiresorptives differs from ALN by sparing bone formation, potentially via uncoupling bone formation from resorption.


Asunto(s)
Enfermedades Óseas Metabólicas/tratamiento farmacológico , Huesos/efectos de los fármacos , Catepsina K/antagonistas & inhibidores , Alendronato/uso terapéutico , Animales , Compuestos de Bifenilo/uso terapéutico , Densidad Ósea , Resorción Ósea , Densitometría , Relación Dosis-Respuesta a Droga , Estradiol/farmacología , Femenino , Fémur/efectos de los fármacos , Osteón/efectos de los fármacos , Conejos
20.
J Bone Miner Res ; 26(5): 1012-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21542004

RESUMEN

Therapeutic enhancement of fracture healing would help to prevent the occurrence of orthopedic complications such as nonunion and revision surgery. Sclerostin is a negative regulator of bone formation, and treatment with a sclerostin monoclonal antibody (Scl-Ab) results in increased bone formation and bone mass in animal models. Our objective was to investigate the effects of systemic administration of Scl-Ab in two models of fracture healing. In both a closed femoral fracture model in rats and a fibular osteotomy model in cynomolgus monkeys, Scl-Ab significantly increased bone mass and bone strength at the site of fracture. After 10 weeks of healing in nonhuman primates, the fractures in the Scl-Ab group had less callus cartilage and smaller fracture gaps containing more bone and less fibrovascular tissue. These improvements at the fracture site corresponded with improvements in bone formation, bone mass, and bone strength at nonfractured cortical and trabecular sites in both studies. Thus the potent anabolic activity of Scl-Ab throughout the skeleton also was associated with an anabolic effect at the site of fracture. These results support the potential for systemic Scl-Ab administration to enhance fracture healing in patients.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Densidad Ósea/efectos de los fármacos , Fracturas del Fémur/fisiopatología , Curación de Fractura/efectos de los fármacos , Glicoproteínas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Diáfisis/efectos de los fármacos , Diáfisis/patología , Diáfisis/fisiopatología , Modelos Animales de Enfermedad , Fémur/efectos de los fármacos , Fémur/patología , Fémur/fisiopatología , Peroné/efectos de los fármacos , Peroné/patología , Peroné/fisiopatología , Glicoproteínas/inmunología , Péptidos y Proteínas de Señalización Intercelular , Macaca fascicularis , Masculino , Tamaño de los Órganos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteotomía , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA