Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
J Exp Med ; 192(4): 463-74, 2000 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-10952716

RESUMO

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


Assuntos
Densidade Óssea/fisiologia , Calcinose/fisiopatologia , Glicoproteínas/metabolismo , Osteoclastos/metabolismo , Osteopetrose/metabolismo , Osteoporose/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fosfatase Ácida/metabolismo , Animais , Aorta/patologia , Western Blotting , Células CHO , Catepsina K , Catepsinas/metabolismo , Cricetinae , Fêmur/anatomia & histologia , Fêmur/diagnóstico por imagem , Fêmur/metabolismo , Glicoproteínas/genética , Humanos , Imuno-Histoquímica , Hibridização In Situ , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , Osteoclastos/ultraestrutura , Osteopetrose/genética , Osteoporose/genética , Osteoprotegerina , Radiografia , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão/metabolismo , Fosfatase Ácida Resistente a Tartarato
2.
J Cell Biol ; 145(3): 527-38, 1999 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-10225954

RESUMO

Osteoprotegerin (OPG) and OPG-ligand (OPGL) potently inhibit and stimulate, respectively, osteoclast differentiation (Simonet, W.S., D.L. Lacey, C.R. Dunstan, M. Kelley, M.-S. Chang, R. Luethy, H.Q. Nguyen, S. Wooden, L. Bennett, T. Boone, et al. 1997. Cell. 89:309-319; Lacey, D.L., E. Timms, H.-L. Tan, M.J. Kelley, C.R. Dunstan, T. Burgess, R. Elliott, A. Colombero, G. Elliott, S. Scully, et al. 1998. Cell. 93: 165-176), but their effects on mature osteoclasts are not well understood. Using primary cultures of rat osteoclasts on bone slices, we find that OPGL causes approximately sevenfold increase in total bone surface erosion. By scanning electron microscopy, OPGL-treated osteoclasts generate more clusters of lacunae on bone suggesting that multiple, spatially associated cycles of resorption have occurred. However, the size of individual resorption events are unchanged by OPGL treatment. Mechanistically, OPGL binds specifically to mature OCs and rapidly (within 30 min) induces actin ring formation; a marked cytoskeletal rearrangement that necessarily precedes bone resorption. Furthermore, we show that antibodies raised against the OPGL receptor, RANK, also induce actin ring formation. OPGL-treated mice exhibit increases in blood ionized Ca++ within 1 h after injections, consistent with immediate OC activation in vivo. Finally, we find that OPG blocks OPGL's effects on both actin ring formation and bone resorption. Together, these findings indicate that, in addition to their effects on OC precursors, OPGL and OPG have profound and direct effects on mature OCs and indicate that the OC receptor, RANK, mediates OPGL's effects.


Assuntos
Reabsorção Óssea/metabolismo , Proteínas de Transporte/farmacologia , Glicoproteínas/farmacologia , Glicoproteínas de Membrana/farmacologia , Osteoclastos/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares , Actinas/metabolismo , Animais , Osso e Ossos/química , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Proteínas de Transporte/metabolismo , Bovinos , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/metabolismo , Glicoproteínas/metabolismo , Células-Tronco Hematopoéticas/citologia , Hipercalcemia/induzido quimicamente , Hipercalcemia/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos , Microscopia Eletrônica de Varredura , Osteoclastos/citologia , Osteoclastos/ultraestrutura , Osteoporose/metabolismo , Osteoprotegerina , Ligante RANK , Ratos , Receptor Ativador de Fator Nuclear kappa-B , Receptores do Fator de Necrose Tumoral/metabolismo
3.
Int J Biochem Cell Biol ; 39(6): 1077-81, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17174136

RESUMO

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


Assuntos
Ligante RANK/genética , Ligante RANK/metabolismo , Sequência de Aminoácidos , Animais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/prevenção & controle , Perfilação da Expressão Gênica , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Osteoprotegerina/metabolismo , Ligante RANK/antagonistas & inibidores , Homologia de Sequência de Aminoácidos
4.
Cancer Res ; 56(17): 4063-70, 1996 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-8752180

RESUMO

The molecular mechanisms by which human cancer cells spread to bone are largely unexplored. The process likely involves cell adhesion molecules (CAMs) that are responsible for homophilic and heterophilic cell-cell interactions. One relevant CAM may be the calcium-dependent transmembrane glycoprotein E-cadherin. To investigate the involvement of E-cadherin in breast cancer metastasis to bone, we used an in vivo model in which osteolytic bone metastases preferentially occur after injections of cancer cells directly into the arterial circulation through the left ventricle of the hearts of nude mice. We have found that E-cadherin-negative human breast cancer cells MDA-MB-231 (MDA-231) develop radiographically detectable multiple osteolytic bone metastases and cachexia in this model. However, MDA-231 breast cancer cells that were transfected with E-cadherin cDNA showed a dramatically impaired capacity to form osteolytic metastases and induce cachexia. Histological and histomorphometrical analyses of bones of mice bearing mock-transfected MDA-231 revealed aggressive metastatic tumor, whereas metastatic tumor burden was significantly decreased in the bones of mice bearing E-cadherin-expressing MDA-231. Nude mice bearing E-cadherin-transfected MDA-231 breast cancer cells survived longer than mice bearing mock-transfected MDA-231 breast cancer cells. Anchorage-dependent and -independent growth in culture and tumor enlargement in the mammary fat pad of nude mice were unchanged between mock-transfected and E-cadherin-expressing MDA-231, suggesting that these differences in metastatic behavior are not due to an impairment of cell growth and tumor-igenicity. Our results show the suppressive effects of E-cadherin expression on bone metastasis by circulating breast cancer cells and suggest that the modulation of expression of this CAM may reduce the destructive effects of breast cancer cells on bone.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/fisiologia , Osteólise/metabolismo , Osteólise/patologia , Animais , Neoplasias Ósseas/patologia , Caquexia/etiologia , Caquexia/metabolismo , Caderinas/biossíntese , Caderinas/genética , Adesão Celular/fisiologia , DNA Complementar/genética , Feminino , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Células Neoplásicas Circulantes/patologia , Transfecção , Células Tumorais Cultivadas
5.
Cancer Res ; 61(11): 4432-6, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389072

RESUMO

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


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/secundário , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Glicoproteínas/farmacologia , Osteólise/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Neoplasias Ósseas/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Transformada , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Camundongos Nus , Osteoprotegerina , Receptores Citoplasmáticos e Nucleares , Receptores do Fator de Necrose Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cancer Res ; 57(15): 3194-9, 1997 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9242449

RESUMO

Humoral hypercalcemia of malignancy results from the effects of tumor-produced factors on bone, kidney, and intestine that disrupt normal calcium homeostasis. Although parathyroid hormone-related protein (PTHrP) is a major mediator of the syndrome, tumors also produce other hypercalcemic factors, such as tumor necrosis factor (TNF), which may modulate the effects of PTHrP. It has been postulated that TNF may counteract the stimulatory effects of PTHrP on bone formation. To examine the effects of TNF on PTHrP-induced changes in calcium and bone metabolism, a murine tumor model of hypercalcemia was used. Nude mice were inoculated with Chinese hamster ovarian (CHO) cells expressing human TNF (CHO/TNF) or nontransfected CHO cells (CHO/-) and further treated with injections of human PTHrP(1-34) or vehicle. The effects of TNF, PTHrP, and the combination of the two factors on blood ionized calcium, osteoclast recruitment, and bone histomorphometry were evaluated. Mice bearing CHO/TNF tumors that were injected with PTHrP had significantly higher calcium concentrations, increased committed osteoclast progenitors, and mature osteoclasts as well as enhanced bone resorption compared with mice bearing CHO/TNF tumors injected with vehicle or those bearing CHO/- tumors injected with PTHrP or vehicle. A 2-fold increase in new woven bone formed in the calvaria at sites of previous bone resorption was observed in CHO/TNF mice treated with PTHrP. Bone formation rates in the vertebrae were similar in both CHO/- and CHO/TNF mice treated with PTHrP. These data demonstrate that the hypercalcemic effects of PTHrP are enhanced by TNF and that this effect is due to the increased production of committed osteoclast precursors with a subsequent increase in osteoclastic bone resorption. Furthermore, PTHrP caused a coupled increase in osteoclastic bone resorption and new bone formation that was not inhibited by TNF. These findings highlight the complex interactions that may occur between tumor-produced factors on bone that result in malignancy-associated hypercalcemia and suggest that TNF may not be responsible for the decreased bone formation seen in some patients with this condition.


Assuntos
Reabsorção Óssea/metabolismo , Hipercalcemia/metabolismo , Proteínas/farmacologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Reabsorção Óssea/induzido quimicamente , Células CHO , Calcificação Fisiológica , Cálcio/sangue , Cricetinae , Humanos , Hipercalcemia/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Osteoclastos/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo , Crânio/efeitos dos fármacos , Crânio/fisiologia , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/fisiologia , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/genética
7.
Cancer Res ; 60(4): 783-7, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10706080

RESUMO

Osteoprotegerin (OPG), a novel, secreted tumor necrosis factor receptor family member that inhibits osteoclast formation and activity was examined for its activity in a syngeneic tumor model of humoral hypercalcemia of malignancy. Normal mice bearing Colon-26 tumors develop increases in both parathyroid hormone-related protein (PTHrP) expression and plasma PTHrP, marked hypercalcemia, and increased bone resorption. OPG, given either at the onset of hypercalcemia or after it had occurred, blocked tumor-induced increases in bone resorption and hypercalcemia and rapidly normalized blood ionized calcium. In tumor-bearing mice, OPG treatments reduced osteoclast activity from approximately 2-fold above normal into the subphysiological range but had no effects on tumor size, tumor-induced cachexia, or PTHrP levels. The potent effects of OPG in this humoral hypercalcemia of malignancy model suggest a potential therapeutic role for OPG in the prevention and treatment of this disorder.


Assuntos
Glicoproteínas/uso terapêutico , Hipercalcemia/prevenção & controle , Neoplasias Experimentais/metabolismo , Receptores Citoplasmáticos e Nucleares , Receptores do Fator de Necrose Tumoral/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Hipercalcemia/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Osteoclastos/efeitos dos fármacos , Osteoprotegerina , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/análise
8.
Bone ; 37(1): 87-95, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15869920

RESUMO

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


Assuntos
Osso e Ossos/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Glicoproteínas/farmacologia , Ovariectomia , Teriparatida/farmacologia , Animais , Osso e Ossos/química , Osso e Ossos/patologia , Cálcio/análise , Quimioterapia Combinada , Microanálise por Sonda Eletrônica , Fêmur/química , Fêmur/efeitos dos fármacos , Fêmur/patologia , Glicoproteínas/uso terapêutico , Humanos , Minerais/análise , Minerais/química , Osteoporose/tratamento farmacológico , Osteoprotegerina , Ratos , Receptores Citoplasmáticos e Nucleares/uso terapêutico , Receptores do Fator de Necrose Tumoral/uso terapêutico , Teriparatida/uso terapêutico
9.
Clin Cancer Res ; 7(10): 2977-83, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11595685

RESUMO

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


Assuntos
Glicoproteínas/sangue , Neoplasias da Próstata/patologia , Receptores Citoplasmáticos e Nucleares/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Ósseas/sangue , Neoplasias Ósseas/secundário , Colágeno/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/patologia , Osteoprotegerina , Fragmentos de Peptídeos/sangue , Hiperplasia Prostática/sangue , Hiperplasia Prostática/patologia , Neoplasias da Próstata/sangue , Receptores do Fator de Necrose Tumoral
10.
J Bone Miner Res ; 8(2): 139-45, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7680183

RESUMO

An automated image analysis method is described for measurement of osteoclasts and resorbing surface in calcified bone. Osteoclasts, osteoid, and mineralized bone were measured in a single section, reacted for acid phosphatase activity, and then stained with orange G and light green stain. Three images were acquired of each field with a monochromatic camera at illuminating wavelengths of 635, 540, and 480 nm (selected using a stage monochromator). These wavelengths were chosen according to the absorption spectra of the different image components to maximize absorption differences between osteoclasts (red), mineralized bone (blue/green), and osteoid (orange). These components were then discriminated according to operator-defined ranges of color density (mineralized bone) or color fraction (osteoid and osteoclasts). A gray level coded segmented image was produced, from which was determined the area, perimeter, and number of each component and the length of contact zones with the marrow and between these components. The method was evaluated by twice measuring 10 bone sections from patients with end-stage liver failure awaiting liver transplantation. The method was quite reproducible, with coefficients of variation varying between 4% for bone volume (% tissue volume) and 22% for osteoid surface (% bone surface). The sections were also measured using a previously established semiautomated method. Coefficients of variation between methods were higher varying between 4% for bone volume (% tissue volume) and 56% for osteoid volume (% bone volume). The automated method gave a substantial time saving compared to the semiautomated method. An interactive technique was used in adjacent sections to evaluate tetracycline labeling and osteoblast surfaces.


Assuntos
Reabsorção Óssea/patologia , Processamento de Imagem Assistida por Computador , Osteoclastos/química , Adolescente , Adulto , Biópsia , Densidade Óssea , Feminino , Humanos , Hepatopatias/patologia , Masculino , Pessoa de Meia-Idade , Espectrofotometria Ultravioleta , Coloração e Rotulagem
11.
J Bone Miner Res ; 14(9): 1478-85, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10469275

RESUMO

Osteoprotegerin (OPG) is a secreted protein that inhibits osteoclast formation and activity and appears to be a critical regulator of bone mass and metabolism. In the current study, mice were challenged with various cytokines and hormones (interleukin-1beta, tumor necrosis factor-alpha, parathyroid hormone, parathyroid hormone-related protein, and 1alpha,25-dihydroxyvitamin D3) that are known to increase bone resorption and cause hypercalcemia and treated concurrently with either a recombinant chimeric Fc fusion form of human OPG, with enhanced biological activity (cOPG) (2.5 mg/kg/day) or vehicle. Mice receiving these bone-resorbing factors became hypercalcemic by day 3 after commencing treatment and had increased bone resorption as evidenced by elevated osteoclast numbers on day 5. Concurrent cOPG treatment prevented hypercalcemia (p < 0.05) and maintained osteoclast numbers in the normal range (p < 0.001). The demonstration that cOPG can inhibit bone resorption suggests that this molecule may be useful in the treatment of diseases including hyperparathyroidism, humoral hypercalcemia of malignancy, osteoporosis, and inflammatory bone disease, which are characterized, in part, by increases in osteoclastic bone resorption.


Assuntos
Reabsorção Óssea , Calcitriol/farmacologia , Glicoproteínas/farmacologia , Hipercalcemia/prevenção & controle , Interleucina-1/farmacologia , Hormônio Paratireóideo/farmacologia , Proteínas/farmacologia , Receptores Citoplasmáticos e Nucleares , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/efeitos dos fármacos , Cálcio/sangue , Contagem de Células/efeitos dos fármacos , Glicoproteínas/química , Humanos , Isomerismo , Masculino , Camundongos , Osteoclastos/efeitos dos fármacos , Osteoprotegerina , Hormônio Paratireóideo/sangue , Proteína Relacionada ao Hormônio Paratireóideo , Radiografia , Redução de Peso/efeitos dos fármacos
12.
J Bone Miner Res ; 16(2): 348-60, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11204435

RESUMO

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


Assuntos
Glicoproteínas/uso terapêutico , Osteoporose/prevenção & controle , Pós-Menopausa , Receptores Citoplasmáticos e Nucleares/uso terapêutico , Método Duplo-Cego , Feminino , Glicoproteínas/administração & dosagem , Glicoproteínas/efeitos adversos , Humanos , Pessoa de Meia-Idade , Osteoprotegerina , Placebos , Receptores Citoplasmáticos e Nucleares/administração & dosagem , Receptores do Fator de Necrose Tumoral
13.
J Bone Miner Res ; 4(4): 507-14, 1989 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2816500

RESUMO

Bone mineral content (BMC) was measured in the os calcis of 232 normal subjects aged 17-82 years. The mean reproducibility (coefficient of variation) of the measurement was 1.8%. Substantial bone loss occurred between the ages of 20 and 50 years, and in females the menopause was associated with additional bone loss. There was no significant difference in the rate of bone loss in females and males, but the mean BMC was greater at all ages in males than in females. We also compared os calcis BMC with spinal bone mineral density (BMD), measured by quantitative computed tomographic (CT) scanning, in 85 subjects: 33 were normal controls, 19 had osteoporosis defined by the presence of one or more pathological fractures, and in the remainder the CT examination was performed at the patient's request. Os calcis BMC correlated with spinal BMD in both females (r = 0.69, p less than 0.001) and males (r = 0.84, p less than 0.001). However, the os calcis BMC did not reliably predict spine values around the CT "fracture threshold" of 90-100 mg/cm3 and did not correlate with osteoporotic fracture as well as did spinal BMD. It is concluded that measurement of the os calcis BMC is of limited clinical usefulness for the early diagnosis of osteoporosis.


Assuntos
Osso e Ossos/análise , Calcâneo/análise , Minerais/análise , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Estatura , Peso Corporal , Feminino , Fraturas Ósseas/metabolismo , Humanos , Masculino , Menopausa , Pessoa de Meia-Idade , Osteoporose/metabolismo , Valores de Referência , Análise de Regressão , Fatores Sexuais , Coluna Vertebral/análise
14.
J Bone Miner Res ; 15(1): 2-12, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10646108

RESUMO

Although multiple hormones and cytokines regulate various aspects of osteoclast formation, the final two effectors are osteoprotegerin ligand (OPG-L)/osteoclast differentiation factor (ODF), a recently cloned member of the tumor necrosis factor superfamily, and macrophage colony-stimulating factor. OPG-L/ODF is produced by osteoblast lineage cells and exerts its biological effects through binding to its receptor, osteoclast differentiation and activation receptor (ODAR)/receptor activator of NF-kappa B (RANK), on osteoclast lineage cells, in either a soluble or a membrane-bound form, the latter of which requires cell-to-cell contact. Binding results in rapid differentiation of osteoclast precursors in bone marrow to mature osteoclasts and, at higher concentrations, in increased functional activity and reduced apoptosis of mature osteoclasts. The biological activity of OPG-L/ODF is neutralized by binding to osteoprotegerin (OPG)/osteoclastogenesis inhibitory factor (OCIF), a member of the TNF-receptor superfamily that also is secreted by osteoblast lineage cells. The biological importance of this system is underscored by the induction in mice of severe osteoporosis by targeted ablation of OPG/OCIF and by the induction of osteopetrosis by targeted ablation of OPG-L/ODF or overexpression of OPG/OCIF. Thus, osteoclast formation may be determined principally by the relative ratio of OPG-L/ODF to OPG/OCIF in the bone marrow microenvironment, and alterations in this ratio may be a major cause of bone loss in many metabolic disorders, including estrogen deficiency and glucocorticoid excess. That changes in but two downstream cytokines mediate the effects of large numbers of upstream hormones and cytokines suggests a regulatory mechanism for osteoclastogenesis of great efficiency and elegance.


Assuntos
Reabsorção Óssea/fisiopatologia , Proteínas de Transporte/metabolismo , Glicoproteínas/fisiologia , Glicoproteínas de Membrana/metabolismo , Receptores Citoplasmáticos e Nucleares , Animais , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicoproteínas/uso terapêutico , Humanos , Osteoclastos/citologia , Osteoprotegerina , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores do Fator de Necrose Tumoral
15.
J Bone Miner Res ; 14(6): 953-9, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10352104

RESUMO

There are no universally accepted agents that will substantially increase bone mass in osteoporotic patients. A number of peptides important in normal bone formation, such as members of the transforming growth factor-beta superfamily, are not satisfactory for this purpose either because their beneficial effects are predominantly local or there is systemic toxicity associated with their administration. We have examined the effects of exogenous fibroblast growth factor-1 and -2 (FGF-1 and FGF-2) on bone in vivo, since FGFs have been shown recently to be essential for normal skeletal development. FGF-1 was injected daily (0.2 mg/kg intravenously) for 28 days into the tail vein of adult female rats immediately following and 6 months after sham operation or ovariectomy (OVX). In rats treated immediately post-OVX, OVX produced more than a 30% decrease in tibial bone density, which was prevented by FGF-1 and estrogen. However, FGF-1 also had an anabolic effect. In sham-operated rats, FGF-1 increased bone density to 2-fold, whereas estrogen had no effect. In rats 6 months post-OVX, severe bone loss and disruption of trabecular microarchitecture occurred similar to that seen in patients with severe osteoporosis. In these rats, administration of FGF-1 induced extensive new woven bone formation with new trabecular-like structures filling much of the marrow spaces, and bone density in the tibial metaphysis increased 3-fold. FGF-1 and FGF-2 were also administered subcutaneously over the calvaria of mice in doses of 2-2000 microg/day for 3 days and shown to produce substantial increases in bone formation when examined morphologically. Thus, we conclude that both local and systemic FGF-1 increases new bone formation and bone density, and systemic FGF-1 also appears to restore bone microarchitecture and prevent bone loss associated with estrogen-withdrawal.


Assuntos
Desenvolvimento Ósseo/efeitos dos fármacos , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Osteoporose Pós-Menopausa/prevenção & controle , Ovário/fisiologia , Animais , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Humanos , Injeções , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Ovariectomia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/uso terapêutico , Crânio/efeitos dos fármacos
16.
Endocrinology ; 141(12): 4768-76, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11108292

RESUMO

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


Assuntos
Proteínas de Transporte/genética , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/genética , Glicoproteínas de Membrana/genética , Osteoblastos/citologia , Osteoclastos/citologia , Receptores Citoplasmáticos e Nucleares/genética , Células Estromais/citologia , Animais , Células da Medula Óssea/fisiologia , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Humanos , Cinética , Camundongos , Osteoprotegerina , Ligante RANK , RNA Mensageiro/análise , Receptor Ativador de Fator Nuclear kappa-B , Receptores do Fator de Necrose Tumoral , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/fisiologia
17.
Endocrinology ; 140(9): 4367-70, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10465311

RESUMO

The identity of the paracrine mediator(s) of the antiresorptive action of estrogen on bone cells is controversial. Osteoprotegerin (OPG) was recently identified as a soluble member of the tumor necrosis factor (TNF) receptor (TNF-R) superfamily that is secreted by osteoblast lineage cells and acts by binding to and neutralizing its cognate ligand, OPG-L, a required factor for osteoclastogenesis. OPG prevents bone loss when administered to ovariectomized rats, induces osteoporosis when ablated in knock-out mice, and induces osteopetrosis when overexpressed in transgenic mice. In conditionally immortalized, human osteoblastic hFOB/ER-3 and hFOB/ER-9 cell lines containing physiological concentrations of approximately 800 and approximately 8,000 functional estrogen receptors (ER)/nucleus, respectively, we found that 17beta-estradiol dose- and time-dependently increased OPG mRNA and protein levels to maximal levels of 370% and 320%, respectively (P < 0.001); co-treatment with the "pure" antiestrogen ICI 182,780 abrogated these effects completely. 17beta-Estradiol also dose-dependently increased OPG mRNA and protein levels in normal human osteoblasts with approximately 400 ER/nucleus by 60% and 73%, respectively. Thus, estrogen enhancement of OPG secretion by osteoblastic cells may play a major role in the antiresorptive action of estrogen on bone.


Assuntos
Estradiol/farmacologia , Expressão Gênica/efeitos dos fármacos , Glicoproteínas/biossíntese , Glicoproteínas/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Receptores Citoplasmáticos e Nucleares , Células Cultivadas , Relação Dose-Resposta a Droga , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Fulvestranto , Humanos , Osteoblastos/metabolismo , Osteoprotegerina , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral , Fatores de Tempo
18.
Endocrinology ; 140(10): 4382-9, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10499489

RESUMO

Osteoporosis is a serious complication of systemic glucocorticoid use. However, while glucocorticoids increase bone resorption in vitro and in vivo, the mechanism(s) of this effect are at present unclear. Recent studies have identified the osteoprotegerin (OPG) ligand (OPG-L) as the final effector of osteoclastogenesis, an action that is opposed by the soluble neutralizing receptor, OPG. Thus, we assessed glucocorticoid regulation of OPG and OPG-L in various human osteoblastic lineage cells using Northern analysis, RT-PCR, and ELISA. Dexamethasone inhibited constitutive OPG messenger RNA (mRNA) steady-state levels by 70-90% in primary (MS) and immortalized stromal cells (hMS), primary trabecular osteoblasts (hOB), immortalized fetal osteoblasts (hFOB), and osteosarcoma cells (MG-63). In hFOB cells, dexamethasone inhibited constitutive OPG mRNA steady-state levels in a dose- and time-dependent fashion by 90%, and also suppressed cytokine-stimulated OPG mRNA steady-state levels. Dexamethasone-induced inhibition of OPG mRNA levels was not affected by the protein synthesis inhibitor, cycloheximide, and was shown to be due to inhibition of OPG gene transcription using a nuclear run-on assay. Moreover, dexamethasone also dose dependently (10(-10) M-10(-7) M) inhibited constitutive OPG protein concentrations in the conditioned medium of hFOB cells from 2.59 +/- 0.02 ng/ml (control) to 0.30 +/- 0.01 ng/ml (88% inhibition; P < 0.001 by ANOVA). Concurrently, dexamethasone stimulated OPG-L mRNA steady-state levels in MS and hFOB cells by 2- and 4-fold, respectively. Treatment of murine marrow cultures with conditioned medium harvested from dexamethasone-treated MG-63 cells increased tartrate-resistant acid phosphatase (TRAP) activity by 54% (P < 0.005) compared with medium harvested from control-treated cells (in the presence of OPG-L and macrophage colony-stimulating factor). Moreover, dexamethasone (10(-8) M) promoted osteoclast formation in vitro, as assessed by a 2.5-fold increase of TRAP activity in cell lysates (P < 0.001) and the appearance of TRAP-positive multinucleated cells. Our data are thus consistent with the hypothesis that glucocorticoids promote osteoclastogenesis by inhibiting OPG and concurrently stimulating OPG-L production by osteoblastic lineage cells, thereby enhancing bone resorption.


Assuntos
Proteínas de Transporte/metabolismo , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Glicoproteínas/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Osteoblastos/metabolismo , Receptores Citoplasmáticos e Nucleares , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cicloeximida/farmacologia , Glucocorticoides/efeitos adversos , Glicoproteínas/genética , Homeostase/efeitos dos fármacos , Humanos , Osteoclastos/citologia , Osteoporose/induzido quimicamente , Osteoprotegerina , Comunicação Parácrina/fisiologia , Inibidores da Síntese de Proteínas/farmacologia , Ligante RANK , RNA Mensageiro/metabolismo , Receptor Ativador de Fator Nuclear kappa-B , Receptores do Fator de Necrose Tumoral
19.
Endocrinology ; 142(10): 4295-304, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11564687

RESUMO

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


Assuntos
Doenças Ósseas Metabólicas/tratamento farmacológico , Glicoproteínas/farmacologia , Fragmentos de Peptídeos/farmacologia , Teriparatida/farmacologia , Animais , Densidade Óssea/efeitos dos fármacos , Doenças Ósseas Metabólicas/fisiopatologia , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Glicoproteínas/uso terapêutico , Osteoprotegerina , Ovariectomia , Fragmentos de Peptídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/uso terapêutico , Receptores do Fator de Necrose Tumoral , Teriparatida/análogos & derivados , Teriparatida/uso terapêutico
20.
J Clin Endocrinol Metab ; 55(6): 1066-72, 1982 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7130337

RESUMO

Quantitative bone histology was studied in 23 patients with malignant hypercalcemia (MH) due to carcinoma (16) or immunoproliferative disease (7). Plasma calcium was 3.37 +/- 0.47 (mean +/- SD) mmol/liter. Bone resorbing surface (RS) was measured using a sensitive histochemical stain to identify osteoclasts. In the MH patients with carcinoma, the RS was 3.1 +/- 2.6% compared to 1.0 +/- 0.3% in controls (P less than 0.02). In the myeloma patients it was 2.3 +/- 1.7%, and in normocalcemic patients with malignant disease 0.8 +/- 1.1%. RS did not correlate with serum PTH, and several high RS values were associated with undetectable PTH. RS correlated with forming surface (FS) in MH patients (r = 0.44, P less than 0.05) and controls (r = 0.68, P less than 0.005), but there was a greater RS relative to FS in MH patients than in controls (P less than 0.005). "Excess" RS in the MH patients was calculated by subtracting the RS accounted for by the measured FS, using the relationship defined by the controls. Bone loss, as reflected in urinary calcium excretion, correlated weakly with excess RS (r = 0.44, P less than 0.05) but was high even when excess RS was zero. Thus, the histological findings do not account for the bone loss, and additional resorption around bone metastases is likely; the results of this study are consistent with a humoral substance produced by the malignant tissue causing generalized bone resorption in addition to bone dissolution around metastases.


Assuntos
Osso e Ossos/patologia , Hipercalcemia/patologia , Neoplasias/patologia , Adulto , Idoso , Reabsorção Óssea , Feminino , Humanos , Hipercalcemia/etiologia , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Neoplasias/complicações , Osteoclastos/patologia , Análise de Regressão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA