Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
MAbs ; 14(1): 1979447, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34923919

RESUMO

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.


Assuntos
Adenoviridae , Linfócitos T CD8-Positivos/imunologia , Vacinas contra a SAIDS , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia/imunologia , Animais , Avaliação de Medicamentos , Macaca fascicularis , Masculino , Vacinas contra a SAIDS/genética , Vacinas contra a SAIDS/imunologia , Vacinas contra a SAIDS/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/genética
2.
Regul Toxicol Pharmacol ; 81: 212-222, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27569204

RESUMO

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.


Assuntos
Anticorpos Monoclonais/toxicidade , Neoplasias/induzido quimicamente , Animais , Anticorpos Monoclonais/administração & dosagem , Testes de Carcinogenicidade , Relação Dose-Resposta a Droga , Humanos , Camundongos , Ratos , Medição de Risco
3.
J Orthop Res ; 34(1): 72-80, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26178170

RESUMO

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.


Assuntos
Compostos de Bifenilo/uso terapêutico , Calo Ósseo/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Consolidação da Fratura/efeitos dos fármacos , Alendronato/farmacologia , Alendronato/uso terapêutico , Animais , Compostos de Bifenilo/farmacologia , Feminino , Osteotomia , Coelhos , Distribuição Aleatória , Ulna
4.
Bone ; 66: 72-81, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24928497

RESUMO

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.


Assuntos
Calo Ósseo/patologia , Calo Ósseo/fisiopatologia , Calcificação Fisiológica , Catepsina K/deficiência , Fraturas do Fêmur/patologia , Fraturas do Fêmur/fisiopatologia , Animais , Fenômenos Biomecânicos , Remodelação Óssea , Catepsina K/metabolismo , Contagem de Células , Feminino , Fraturas do Fêmur/diagnóstico por imagem , Consolidação da Fratura , Deleção de Genes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Osteoblastos/patologia , Osteoclastos/patologia , Radiografia
5.
J Endocrinol ; 212(2): 179-86, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22062085

RESUMO

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.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Reabsorção Óssea/prevenção & controle , Fenofibrato/uso terapêutico , Osteoporose Pós-Menopausa/tratamento farmacológico , PPAR alfa/agonistas , PPAR gama/agonistas , Tiazolidinedionas/efeitos adversos , Adiposidade/efeitos dos fármacos , Animais , Biomarcadores/sangue , Densidade Óssea/efeitos dos fármacos , Conservadores da Densidade Óssea/administração & dosagem , Reabsorção Óssea/induzido quimicamente , Reabsorção Óssea/etiologia , Osso e Ossos/química , Osso e Ossos/efeitos dos fármacos , Colágeno Tipo I/sangue , Quimioterapia Combinada , Feminino , Fenofibrato/administração & dosagem , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/uso terapêutico , Hipolipemiantes/administração & dosagem , Hipolipemiantes/efeitos adversos , Hipolipemiantes/uso terapêutico , Osteocalcina/sangue , Osteoporose Pós-Menopausa/sangue , Osteoporose Pós-Menopausa/fisiopatologia , Ovariectomia , Peptídeos/sangue , Pioglitazona , Distribuição Aleatória , Ratos , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/uso terapêutico
6.
Bone ; 49(3): 376-86, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21658483

RESUMO

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.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiologia , Estrogênios Conjugados (USP)/farmacologia , Estrogênios/farmacologia , Indóis/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Animais , Biomarcadores/metabolismo , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/anatomia & histologia , Feminino , Fêmur/anatomia & histologia , Fêmur/efeitos dos fármacos , Fêmur/fisiologia , Humanos , Vértebras Lombares/anatomia & histologia , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/fisiologia , Tamanho do Órgão , Ovariectomia , Ratos , Ratos Sprague-Dawley , Tíbia/anatomia & histologia , Tíbia/efeitos dos fármacos , Tíbia/fisiologia , Útero/anatomia & histologia , Útero/efeitos dos fármacos
7.
J Bone Miner Res ; 26(5): 1012-21, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21542004

RESUMO

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.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Densidade Óssea/efeitos dos fármacos , Fraturas do Fêmur/fisiopatologia , Consolidação da Fratura/efeitos dos fármacos , Glicoproteínas/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Diáfises/efeitos dos fármacos , Diáfises/patologia , Diáfises/fisiopatologia , Modelos Animais de Doenças , Fêmur/efeitos dos fármacos , Fêmur/patologia , Fêmur/fisiopatologia , Fíbula/efeitos dos fármacos , Fíbula/patologia , Fíbula/fisiopatologia , Glicoproteínas/imunologia , Peptídeos e Proteínas de Sinalização Intercelular , Macaca fascicularis , Masculino , Tamanho do Órgão/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteotomia , Ratos , Ratos Sprague-Dawley
8.
J Bone Miner Res ; 26(2): 252-62, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20734451

RESUMO

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.


Assuntos
Doenças Ósseas Metabólicas/tratamento farmacológico , Osso e Ossos/efeitos dos fármacos , Catepsina K/antagonistas & inibidores , Alendronato/uso terapêutico , Animais , Compostos de Bifenilo/uso terapêutico , Densidade Óssea , Reabsorção Óssea , Densitometria , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Feminino , Fêmur/efeitos dos fármacos , Ósteon/efeitos dos fármacos , Coelhos
9.
Endocrinology ; 148(6): 2613-21, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17332067

RESUMO

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.


Assuntos
Osso e Ossos/fisiologia , Hormônio Foliculoestimulante/fisiologia , Homeostase , Ovário/fisiologia , Androgênios/farmacologia , Animais , Densidade Óssea/genética , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Feminino , Homeostase/genética , Masculino , Camundongos , Camundongos Knockout , Osteoporose/patologia , Ovariectomia , Ovário/fisiopatologia , Ovário/transplante , Receptores do FSH/genética
10.
Endocrinology ; 148(6): 2778-87, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17379647

RESUMO

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.


Assuntos
Alendronato/uso terapêutico , Reabsorção Óssea/prevenção & controle , Osso e Ossos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Alendronato/administração & dosagem , Animais , Densidade Óssea/efeitos dos fármacos , Conservadores da Densidade Óssea/uso terapêutico , Osso e Ossos/metabolismo , Quimioterapia Combinada , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Metabolismo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/efeitos dos fármacos , Osteoprotegerina/administração & dosagem , Osteoprotegerina/uso terapêutico , Ovariectomia
11.
J Bone Miner Res ; 22(1): 55-63, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17014384

RESUMO

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.


Assuntos
Alendronato/farmacologia , Osso e Ossos/fisiologia , Osteoprotegerina/farmacologia , Hormônio Paratireóideo/farmacologia , Coluna Vertebral/fisiologia , Animais , Osso e Ossos/efeitos dos fármacos , Feminino , Fêmur/efeitos dos fármacos , Fêmur/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Coluna Vertebral/efeitos dos fármacos
12.
J Rheumatol ; 33(6): 1167-75, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16755666

RESUMO

OBJECTIVE: Prostaglandins are important mediators in bone metabolism and in pathologies such as rheumatoid arthritis and osteoarthritis. We investigated the roles of cyclooxygenases (COX) and prostaglandin receptors in the accumulation of osteoprotegerin (OPG) in the supernatants of human osteoblasts in culture. METHODS: Three different cellular models were used, the human osteosarcoma cell lines MG-63 and Saos-2, and primary cultures of human osteoblasts. OPG concentrations were determined by ELISA. RESULTS: RT-PCR analysis showed that, like primary human osteoblasts, MG-63 cells express DP, EP4, FP, IP, and TP receptors, whereas the Saos-2 cells lack IP. Concentration of OPG was highest in MG-63 cell supernatants (36 +/- 12.5 ng/ml), followed by human osteoblasts (12.77 +/- 2.2 ng/ml) and Saos-2 (3.6 +/- 0.76 ng/ml). COX inhibitors did not alter these values. Prostaglandin E2 and BW 245C (a synthetic DP receptor agonist) decreased OPG in the supernatants of human osteoblasts but not in immortalized cell lines. These effects were concentration-dependent and were inhibited by EP4 and DP receptor antagonists. Fluprostenol, an FP receptor agonist, increased the accumulation of OPG in MG-63 but not in primary human osteoblasts or Saos-2. CONCLUSION: Our results show that activation of EP4 or DP receptors decreased the accumulation of OPG in supernatants of osteoblasts in culture, and suggest that these receptors could be interesting pharmacological targets in bone diseases. They also demonstrate important differences between primary osteoblasts and immortalized cell lines, both in the distribution and in the effects mediated by prostaglandin receptors.


Assuntos
Glicoproteínas/metabolismo , Osteoblastos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Humanos , Hidantoínas/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteoprotegerina , Prostaglandina-Endoperóxido Sintases/metabolismo , Prostaglandinas F Sintéticas/farmacologia , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/genética , Receptores do Fator de Necrose Tumoral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Bone Miner Res ; 20(4): 672-81, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15765187

RESUMO

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.


Assuntos
Quimiotaxia , Osteoblastos/metabolismo , Prostaglandina D2/biossíntese , Receptores Imunológicos/fisiologia , Receptores de Prostaglandina/fisiologia , Osso e Ossos/metabolismo , Proteínas de Transporte/metabolismo , AMP Cíclico/metabolismo , Glicoproteínas/metabolismo , Substâncias de Crescimento/farmacologia , Humanos , Hidantoínas/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-1/farmacologia , Glicoproteínas de Membrana/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoprotegerina , Prostaglandina D2/genética , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA