Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
J Exp Med ; 192(4): 463-74, 2000 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-10952716

RESUMEN

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


Asunto(s)
Densidad Ósea/fisiología , Calcinosis/fisiopatología , Glicoproteínas/metabolismo , Osteoclastos/metabolismo , Osteopetrosis/metabolismo , Osteoporosis/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Aorta/patología , Western Blotting , Células CHO , Catepsina K , Catepsinas/metabolismo , Cricetinae , Fémur/anatomía & histología , Fémur/diagnóstico por imagen , Fémur/metabolismo , Glicoproteínas/genética , Humanos , Inmunohistoquímica , Hibridación in Situ , Isoenzimas/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/ultraestructura , Osteopetrosis/genética , Osteoporosis/genética , Osteoprotegerina , Radiografía , Receptores Citoplasmáticos y Nucleares/genética , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes de Fusión/metabolismo , Fosfatasa Ácida Tartratorresistente
2.
J Cell Biol ; 145(3): 527-38, 1999 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-10225954

RESUMEN

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.


Asunto(s)
Resorción Ósea/metabolismo , Proteínas Portadoras/farmacología , Glicoproteínas/farmacología , Glicoproteínas de Membrana/farmacología , Osteoclastos/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares , Actinas/metabolismo , Animales , Huesos/química , Huesos/citología , Huesos/metabolismo , Proteínas Portadoras/metabolismo , Bovinos , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citoesqueleto/metabolismo , Glicoproteínas/metabolismo , Células Madre Hematopoyéticas/citología , Hipercalcemia/inducido químicamente , Hipercalcemia/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos , Microscopía Electrónica de Rastreo , Osteoclastos/citología , Osteoclastos/ultraestructura , Osteoporosis/metabolismo , Osteoprotegerina , Ligando RANK , Ratas , Receptor Activador del Factor Nuclear kappa-B , Receptores del Factor de Necrosis Tumoral/metabolismo
3.
J Clin Invest ; 91(4): 1731-42, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8473513

RESUMEN

IL-1-induced osteoblast IL-6 production represents one possible mechanism by which IL-1 augments bone resorption. In this report, we show that the murine osteoblastic cell line (MC3T3-E1) expresses type 1 IL-1 receptors based on 125I-HrIL1 alpha binding, blocked by type 1 IL-1R antibodies (35F5), and analysis of MC3T3 RNA by reverse transcription (RT)-DNA amplification and Northern analysis. MC3T3 cells do not express detectable type 2 IL-1R mRNA by RT-DNA amplification. IL-1 induces (IL-1 ED50, 0.1 pM) IL-6 production through the type 1 IL-1R as 35F5 antibodies block IL-1-stimulated IL-6 production. Vitamin D3 increases IL-1R expression dose- and metabolite-dependently, with 1,25-(OH)2D3 having the greatest potency, and also enhances IL-1's capacity to stimulate IL-6 production at low IL-1 levels. Both IL-1 and 1,25-(OH)2D3 induce type 1 IL-1R and not type 2 IL-1R upregulation based on ligand binding and RT-DNA amplification. Increased IL-1R expression requires a 5-7-h treatment and is protein/RNA synthesis dependent. These observations imply that IL-1-induced IL-6 production in osteoblasts is mediated by type 1 IL-1Rs and that increased IL-1R expression could play a role in mediating IL-1-induced skeletal responses.


Asunto(s)
Calcitriol/farmacología , Interleucina-1/farmacología , Interleucina-6/metabolismo , Osteoblastos/metabolismo , Receptores de Interleucina-1/fisiología , Células 3T3/ultraestructura , Animales , Anticuerpos/farmacología , Secuencia de Bases , Unión Competitiva , Northern Blotting , Humanos , Interleucina-1/fisiología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , ARN Mensajero/análisis , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Proteínas Recombinantes/farmacología , Regulación hacia Arriba/efectos de los fármacos
4.
Cancer Res ; 61(11): 4432-6, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11389072

RESUMEN

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


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/secundario , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Glicoproteínas/farmacología , Osteólisis/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Neoplasias Óseas/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Transformada , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Desnudos , Osteoprotegerina , Receptores Citoplasmáticos y Nucleares , Receptores del Factor de Necrosis Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 60(4): 783-7, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10706080

RESUMEN

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.


Asunto(s)
Glicoproteínas/uso terapéutico , Hipercalcemia/prevención & control , Neoplasias Experimentales/metabolismo , Receptores Citoplasmáticos y Nucleares , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Hipercalcemia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Osteoclastos/efectos de los fármacos , Osteoprotegerina , Proteína Relacionada con la Hormona Paratiroidea , Proteínas/análisis
6.
Cancer Res ; 58(5): 933-9, 1998 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-9500453

RESUMEN

Keratinocyte growth factor (KGF) stimulates the proliferation and differentiation of epithelial cells including those of the gastrointestinal tract. Although chemotherapeutics and radiation exposure kill rapidly proliferating tumor cells, rapidly dividing normal cells of the host's gastrointestinal tract are also frequently damaged, leading to the clinical condition broadly termed "mucositis." In this report, recombinant human KGF used as a pretreatment in several mouse models of chemotherapy and/or radiation-induced gastrointestinal injury significantly improved mouse survival. Using multiple-dose 5-fluorouracil, methotrexate, and radiation in combination and total body radiation alone models, KGF increased survival by 55% or greater. In the models that used chemotherapy with or without radiation, KGF significantly ameliorated weight loss after injury and accelerated weight gain during recovery. The basis of these systemic benefits appears to be due in part to the trophic effects of the growth factor on the intestinal epithelium because KGF pretreatment caused an increase in measures of mucosal thickness (villus height and crypt depth) that persisted during the course of 5-fluorouracil chemotherapy. Treatment with KGF also afforded a 3.5-fold improvement in crypt survival in the small intestine, suggesting that KGF also has a direct effect on the crypt stem cells. These data indicate that KGF may be therapeutically useful to lessen the intestinal side effects of current cancer therapy regimens.


Asunto(s)
Antineoplásicos/efectos adversos , Factores de Crecimiento de Fibroblastos , Sustancias de Crecimiento/uso terapéutico , Mucosa Intestinal/lesiones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/radioterapia , Traumatismos Experimentales por Radiación/prevención & control , Animales , Femenino , Factor 10 de Crecimiento de Fibroblastos , Factor 7 de Crecimiento de Fibroblastos , Sustancias de Crecimiento/administración & dosificación , Humanos , Enfermedades Intestinales/prevención & control , Mucosa Intestinal/efectos de los fármacos , Ratones , Ratones Desnudos , Neoplasias Experimentales/mortalidad , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Análisis de Supervivencia
7.
Exp Hematol ; 27(9): 1409-17, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10480432

RESUMEN

In this report, we tested whether ectopic overexpression of a cell surface receptor cDNA could be used to explore the physiological roles of that receptor. We generated c-mpl overexpressing animals by reconstituting mice with retroviral vector-transduced bone marrow (BM) cells. We observed that platelet counts in the c-mpl overexpressing mice failed to recover to normal levels and remained at <200 x 10(6)/mL post-transplantation, while platelet numbers in the control mice returned to > 800 x 10(6)/mL by 4 weeks post-transplantation. However, platelet counts in the c-mpl overexpressing mice could be stimulated to normal levels after administration of rhMGDF. No significant changes in peripheral leukocyte counts were observed, although the number of CFU-E, GM-CFC, and CFC-multi were reduced two- to threefold in the BM of the c-mpl overexpressing mice. In addition, enhanced erythropoiesis was observed in the c-mpl overexpressing mice. The mpl receptors on erythroid cells were functional as demonstrated by tyrosine-phosphorylation of mpl receptor on RBC and by in vitro erythroid colony-formation in response to MGDF stimulation, respectively. These results suggested that ectopically expressed mpl receptors competed for ligand in vivo leading to an insufficient amount of circulating thrombopoietin (Tpo) for the development of megakaryocytic lineage. These results further suggest that, in addition to sequestering circulating Tpo, overexpression of the mpl receptor on erythroid progenitors may directly contribute to enhanced erythropoiesis in vivo. Our studies demonstrate that ectopic overexpression of a receptor by retroviral-mediated gene transfer provides an approach to explore the biological roles of novel receptors.


Asunto(s)
Eritropoyesis , Hematopoyesis , Megacariocitos/citología , Proteínas de Neoplasias , Proteínas Proto-Oncogénicas/biosíntesis , Receptores de Citocinas , Animales , Ensayo de Unidades Formadoras de Colonias , ADN Complementario/genética , Vectores Genéticos/genética , Factores de Crecimiento de Célula Hematopoyética/farmacología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/virología , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteínas Proto-Oncogénicas/genética , Provirus/genética , Receptores de Trombopoyetina , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes/farmacología , Retroviridae/genética , Trombopoyetina/farmacología , Trombopoyetina/fisiología , Transfección
8.
J Bone Miner Res ; 14(9): 1478-85, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10469275

RESUMEN

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.


Asunto(s)
Resorción Ósea , Calcitriol/farmacología , Glicoproteínas/farmacología , Hipercalcemia/prevención & control , Interleucina-1/farmacología , Hormona Paratiroidea/farmacología , Proteínas/farmacología , Receptores Citoplasmáticos y Nucleares , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Huesos/diagnóstico por imagen , Huesos/efectos de los fármacos , Calcio/sangre , Recuento de Células/efectos de los fármacos , Glicoproteínas/química , Humanos , Isomerismo , Masculino , Ratones , Osteoclastos/efectos de los fármacos , Osteoprotegerina , Hormona Paratiroidea/sangre , Proteína Relacionada con la Hormona Paratiroidea , Radiografía , Pérdida de Peso/efectos de los fármacos
9.
J Bone Miner Res ; 15(1): 2-12, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10646108

RESUMEN

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.


Asunto(s)
Resorción Ósea/fisiopatología , Proteínas Portadoras/metabolismo , Glicoproteínas/fisiología , Glicoproteínas de Membrana/metabolismo , Receptores Citoplasmáticos y Nucleares , Animales , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicoproteínas/uso terapéutico , Humanos , Osteoclastos/citología , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores del Factor de Necrosis Tumoral
10.
Endocrinology ; 136(6): 2367-76, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7750457

RESUMEN

Interleukin 4 (IL-4) is an immune cytokine that inhibits bone resorption in mice and suppresses osteoclastic cell formation in vitro through an undefined mechanism. In this report, we have established the cellular identity of the IL-4 target cell using a variety of bone marrow/stromal cell coculture methods. Initially, we found that the majority of IL-4's inhibition of osteoclastic cell formation was due to its effect on bone marrow cells, not stromal cells. Consequently, bone marrow macrophages were used as osteoclastic cell progenitors after they had been transiently exposed to IL-4 (48 h), before the addition of stromal cells, 1,25-dihydroxyvitamin D3, and dexamethasone. In this circumstance, IL-4 impaired subsequent osteoclastic cell formation, suggesting that the macrophage may be potentially targeted by many factors known to influence osteoclast formation. Consequently, we discovered that interferon-gamma (IFN gamma), prostaglandin E (PGE), and cell-permeant cAMP analogs also impacted osteoclastic cell formation when used to selectively treat bone marrow macrophages. IFN gamma suppressed osteoclastic cell formation, whereas PGE and cAMP analog treatment led to the formation of significantly enlarged osteoclastic cells. Importantly, PGE antagonized the inhibitory effects of both IL-4 and IFN gamma on the osteoclastic cell-forming potential of bone marrow macrophages. Collectively, these findings establish bone marrow macrophages as osteoclastic cell precursors with the degree of their commitment to the osteoclast pathway sensitive to the effects of soluble mediators, including IL-4, IFN gamma, and PGE.


Asunto(s)
Células de la Médula Ósea , Médula Ósea/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-4/farmacología , Prostaglandinas E/farmacología , Animales , Resorción Ósea/prevención & control , Ensayo de Unidades Formadoras de Colonias , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Técnicas In Vitro , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Modelos Biológicos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos
11.
Endocrinology ; 140(10): 4382-9, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10499489

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Dexametasona/farmacología , Glucocorticoides/farmacología , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Osteoblastos/metabolismo , Receptores Citoplasmáticos y Nucleares , División Celular/efectos de los fármacos , Línea Celular , Cicloheximida/farmacología , Glucocorticoides/efectos adversos , Glicoproteínas/genética , Homeostasis/efectos de los fármacos , Humanos , Osteoclastos/citología , Osteoporosis/inducido químicamente , Osteoprotegerina , Comunicación Paracrina/fisiología , Inhibidores de la Síntesis de la Proteína/farmacología , Ligando RANK , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B , Receptores del Factor de Necrosis Tumoral
12.
Endocrinology ; 142(10): 4295-304, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11564687

RESUMEN

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


Asunto(s)
Enfermedades Óseas Metabólicas/tratamiento farmacológico , Glicoproteínas/farmacología , Fragmentos de Péptidos/farmacología , Teriparatido/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Enfermedades Óseas Metabólicas/fisiopatología , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Glicoproteínas/uso terapéutico , Osteoprotegerina , Ovariectomía , Fragmentos de Péptidos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/uso terapéutico , Receptores del Factor de Necrosis Tumoral , Teriparatido/análogos & derivados , Teriparatido/uso terapéutico
13.
Endocrinology ; 140(9): 4367-70, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10465311

RESUMEN

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.


Asunto(s)
Estradiol/farmacología , Expresión Génica/efectos de los fármacos , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Receptores Citoplasmáticos y Nucleares , Células Cultivadas , Relación Dosis-Respuesta a Droga , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Fulvestrant , Humanos , Osteoblastos/metabolismo , Osteoprotegerina , ARN Mensajero/metabolismo , Receptores del Factor de Necrosis Tumoral , Factores de Tiempo
14.
J Clin Endocrinol Metab ; 85(1): 275-85, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10634399

RESUMEN

We previously reported that keratinocyte growth factor (KGF) is up-regulated by the action of progesterone (P) in the primate endometrium, and we suggested that this protein is a likely mediator of P-dependent stromal-epithelial paracrine interactions in this tissue. At the end of the menstrual cycle, P levels fall, and the abundance of endometrial KGF transcripts decreases approximately 9-fold. In macaques, withdrawal of P induces the luteal-follicular transition (LFT), marked by menstrual sloughing of the functionalis zone and apoptotic regression of the basalis zone. Because KGF levels fall so dramatically during the LFT, we hypothesized that replacement with exogenous KGF during the LFT would prevent some of the endometrial changes seen after P withdrawal. Here we describe two studies of the effects of exogenously administered KGF during the LFT in rhesus macaques. In one experiment we administered KGF systemically to ovariectomized, juvenile rhesus macaques during an LFT induced by hormonal manipulations. KGF had dramatic proliferative effects on the bladder and salivary glands, known targets of KGF, but did not affect cell proliferation in the endometrium or block menstrual sloughing and bleeding. However, KGF strongly inhibited apoptosis in the basalis zone, increased glandular sacculation and folding in this zone, and had a marked trophic effect on the spiral arteries. In the second experiment we installed oviductal catheters in ovariectomized adult rhesus macaques and infused KGF directly into the uterine lumen during a hormonally induced LFT. Again, arteriotrophic, antiapoptotic, and basalis gland sacculation effects were observed in the absence of any effect on cell proliferation. We concluded that although KGF is mitogenic for many epithelial cell types, it does not play this role in the primate endometrium. Its most important roles may be to stimulate spiral artery growth and inhibit glandular apoptosis during the nonfertile menstrual cycle. Because its expression rises coincident with the time of implantation and because spiral arteries are essential to successful establishment of pregnancy, the role of KGF in the fertile menstrual cycle deserves further study.


Asunto(s)
Endometrio/efectos de los fármacos , Factores de Crecimiento de Fibroblastos , Fase Folicular/fisiología , Sustancias de Crecimiento/farmacología , Queratinocitos/efectos de los fármacos , Fase Luteínica/fisiología , Animales , Apoptosis/efectos de los fármacos , Bromodesoxiuridina/farmacología , Cateterismo , Endometrio/citología , Estradiol/sangre , Estradiol/farmacología , Trompas Uterinas/efectos de los fármacos , Trompas Uterinas/fisiología , Femenino , Factor 10 de Crecimiento de Fibroblastos , Factor 7 de Crecimiento de Fibroblastos , Inmunohistoquímica , Macaca mulatta , Ciclo Menstrual/efectos de los fármacos , Ciclo Menstrual/fisiología , Ovariectomía , Péptidos/farmacología
15.
Cell Prolif ; 35 Suppl 1: 78-85, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12139710

RESUMEN

The epithelium of the oral cavity and small intestine of the gastrointestinal tract have a high rate of cell renewal and as such, are sensitive to cytotoxic therapies that kill rapidly dividing cells. Mucositis is a complication of cancer therapy where impairment of the regenerative capacity of the epithelium leads to atrophy, ulceration and a loss of barrier function. Keratinocyte growth factor (KGF) is an epithelial cell-specific growth and differentiation factor that is trophic for the mucosal epithelium of the gastrointestinal tract. In this study, KGF in normal animals caused epithelial thickening in the squamous epithelium of the oral cavity and increased crypt depth and villus height of the small intestine. It also appeared to regulate gene expression in these tissues including that of some antioxidant enzymes and intestinal trefoil protein. KGF has been shown to be efficacious in several preclinical models of mucositis where KGF pretreatment reduced weight loss typically seen during and after the course of therapy and significantly improved survival. At a tissue level KGF reduced atrophy, accelerated regrowth, and decreased ulcer formation of the oral epithelium after irradiation, and improved crypt survival and prevented villus atrophy in the small intestine of irradiated or chemotherapy-treated mice. Preliminary studies suggest that its efficacy may be partly a consequence of the growth and differentiation effect, and also partly due to regulation of the expression of genes that play a role in mucosal protection. These data suggest that KGF may be useful for the prevention or treatment of mucositis in patients treated with regimens of cancer therapy that have gastrointestinal toxicity.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Mucosa Bucal/patología , Estomatitis/tratamiento farmacológico , Estomatitis/patología , Animales , Modelos Animales de Enfermedad , Factor 7 de Crecimiento de Fibroblastos , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología
16.
Bone ; 26(5): 443-9, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10773583

RESUMEN

Osteoprotegerin (OPG) is a recently discovered protein related to the tumor necrosis factor receptor family. It has been shown to inhibit ovariectomy (ovx)-induced resorption in rats and increase bone mineral density in young mice. Tail suspension is a procedure that inhibits bone formation in maturing rodents. This study was designed to quantify OPG's effect on cortical bone formation. Fifty-four mice were assigned to one of five groups (n = 10-11/group). A baseline control group was killed on day 0 of the 10 day study. The remaining groups were: vivarium housed (nonsuspended) control mice receiving 0.3 mg/kg per day OPG; vivarium control mice receiving daily placebo injections; tail-suspended mice receiving 0. 3 mg/kg per day OPG; and tail-suspended mice receiving placebo injections. Tetracycline was administered on days 0 and 8. OPG treatment of tail-suspended mice produced mechanical properties similar to those of placebo-treated, vivarium-housed mice: structural stiffness (8.5%, 20.7%) and elastic (13.9%, 10.1%) and maximum (4.7%, 8.1%) force were increased compared with placebo controls (vivarium, suspended groups). Percent mineral composition was highly significantly greater (p < 0.001 for all comparisons) for OPG-treated mice in the femur, tibia, and humerus, relative to placebo treatment. Matrix mass was also significantly increased in the femur, although not to the same degree as mineral mass. OPG decreased the amount of femoral endocortical resorption compared with the placebo-treated groups for both vivarium (27%) and suspended (24%) mice. Administration of OPG significantly decreased endocortical formation of the tibia. Periosteal bone formation rates were not altered by OPG. OPG-mitigated tail suspension induced osteopenia not by returning bone formation to normal levels, but by inhibiting resorption and increasing percent mineral composition.


Asunto(s)
Enfermedades Óseas Metabólicas/tratamiento farmacológico , Glicoproteínas/farmacología , Suspensión Trasera , Receptores Citoplasmáticos y Nucleares , Animales , Enfermedades Óseas Metabólicas/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoprotegerina , Placebos , Ratas , Receptores del Factor de Necrosis Tumoral
17.
Bone ; 25(3): 255-9, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10495128

RESUMEN

Recent studies have identified osteoprotegerin ligand (OPG-L) as the essential factor required for osteoclastogenesis, and that the effects are prevented by its soluble receptor, osteoprotegerin (OPG). However, there are limited data at present on the regulation of OPG-L expression in human osteoblastic cells by other cytokines. Because interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha, and IL-6 all increase osteoclastogenesis, we assessed whether OPG-L mRNA steady-state levels were regulated by these cytokines in human osteoblastic cells. By northern analysis, IL-1beta (5 nmol/L) and TNF-alpha (9 nmol/L) increased OPG-L mRNA steady-state levels by up to two- to three-fold in normal marrow stromal cells (MS), an immortalized marrow stromal cell line (hMS), and the osteosarcoma cell line, MG-63, whereas IL-6 (2 nmol/L, with or without its soluble receptor) had no effect on OPG-L mRNA levels in any of these cells. IL-1beta and TNF-alpha increased OPG-L mRNA steady-state levels in the normal MS cells and the hMS cell line in a time- and dose-dependent fashion by up to 4.1-fold and up to 2.6-fold, respectively. Our data are thus consistent with the hypothesis that the proinflammatory and bone-resorbing cytokines, IL-1beta and TNF-alpha, but not IL-6, may stimulate osteoclastogenesis by inducing the expression of OPG-L.


Asunto(s)
Proteínas Portadoras/genética , Interleucina-1/farmacología , Interleucina-6/farmacología , Glicoproteínas de Membrana/genética , Osteoblastos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Northern Blotting , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proteínas Portadoras/biosíntesis , Línea Celular Transformada , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1/genética , Interleucina-6/genética , Ligandos , Glicoproteínas de Membrana/biosíntesis , Osteoblastos/metabolismo , Ligando RANK , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
18.
Int J Radiat Oncol Biol Phys ; 40(1): 177-87, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9422575

RESUMEN

PURPOSE: Keratinocyte growth factor (KGF) has potent mitogenic activity on normal epithelial cells and has been found to enhance intestinal crypt cell survival in irradiated mice and to prevent radiation and chemotherapy-induced mucositis in animal models. The purpose of the study reported here is to investigate the effect of recombinant human KGF on the proliferation and survival of human squamous carcinoma cell lines following irradiation. METHODS AND MATERIALS: The level of KGF receptor (KGFR) mRNA in normal Balb/Mk cell line and human head and neck squamous carcinoma cell lines was assessed using a RNase protection assay. The clonogenic assay and MTT assay were used to study the proliferative effects of KGF on human tumor cell lines and Balb/MK cell line in vitro. Effects of KGF on in vivo tumor growth and radiosensitivity were studied in three KGFR-positive human squamous cell carcinoma xenografts (FaDu, Detroit 562 and A431) in nude mice, and a murine KGFR-negative melanoma tumor (B16) in Balb/c mice. RESULTS: Seven of 10 tumor cell lines studied expressed KGFR mRNA. None of these tumor cell lines showed enhanced proliferation when exposed to KGF for 2 days or less. Prolonged exposure to KGF for 7 days or longer resulted in low level stimulation of proliferation in both clonogenic and MTT assays in four of seven KGFR-positive cell lines. Two KGFR-negative cell lines also had a low proliferative response to KGF in a clonogenic assay, but not in the MTT assay. Normal keratinocyte Balb/MK cells, which expressed a moderate level of KGFR mRNA, had a strongly proliferative response to KGF. Its KGF enhancement ratio (KER) of plating efficiency was 24-70 times higher than that of the tumor cells studied (p < 0.001). The KGF-stimulated tumor cell growth was almost completely inhibited by heparin or epidermal growth factor (EGF). There were no significant differences (p > 0.05) in the survival of any of tumor cell lines in the presence or absence of KGF (100 ng/ml) irradiated with doses of 0-15 Gy, and no significant differences (p > 0.05) between the radiobiological parameters D0, Dq, and n number from the SHMT model, alpha, beta, and alpha/beta ratio from the LQ model and SF2 for radiation survival curves for cell lines irradiated in the presence or absence of KGF. Three KGFR-positive human squamous cell carcinoma xenografts in nude mice, and a murine KGFR-negative melanoma tumor in Balb/c mice treated with 1.0 mg/kg of KGF for 3 days grew at the same rate as in untreated mice. CONCLUSION: The recombinant human KGF resulted in little or no stimulation of the proliferation of human head and neck squamous tumor cell lines and did not affect the radiosensitivity of these cell lines in vitro and in vivo. Therefore, KGF may be of clinical value in preventing radiation-induced mucositis and may have the potential to increase the therapeutic index of radiotherapy for treatment of cancers.


Asunto(s)
Carcinoma de Células Escamosas/patología , Supervivencia Celular/efectos de los fármacos , Factores de Crecimiento de Fibroblastos , Sustancias de Crecimiento/farmacología , Receptores de Factores de Crecimiento de Fibroblastos , Animales , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Ensayo de Unidades Formadoras de Colonias , Factor de Crecimiento Epidérmico/farmacología , Femenino , Factor 10 de Crecimiento de Fibroblastos , Factor 7 de Crecimiento de Fibroblastos , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento/metabolismo , Proteínas Recombinantes/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación
19.
Transplantation ; 72(8): 1354-62, 2001 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-11685103

RESUMEN

BACKGROUND: Reactive oxygen species (ROS) and glutathione (GSH) depletion contribute to organ injury after bone marrow transplantation (BMT). Keratinocyte growth factor (KGF) ameliorates graft-versus-host disease (GVHD)-associated organ injury in murine BMT models. METHODS: B10.BR mice received total body irradiation (TBI; day -1) +/- cyclophosphamide (Cy; 120 mg/kg/day i.p., days -3 and -2), then were transplanted on day 0 with C57BL/6 bone marrow + spleen cells as a source of GVHD-causing T cells. KGF (5 mg/kg/day subcutaneously [s.c.]) or saline was given on days -6, -5, and -4. Lung and liver GSH and oxidized GSH disulfide (GSSG) levels were measured on days 0 and 5 and glutathione redox potential (Eh) calculated. Organ malondialdehyde (MDA) was determined on day 5 as an index of ROS-mediated lipid peroxidation. RESULTS: In lung, TBI+BMT oxidized GSH Eh and increased MDA. Cy further oxidized lung GSH Eh. In liver, neither BMT regimen altered GSH redox status or MDA. KGF prevented the decrease in lung GSH after TBI+Cy and decreased lung MDA after both TBI and TBI+Cy. KGF increased liver GSH levels and GSH Eh after TBI and GSH Eh after TBI+Cy. CONCLUSIONS: In murine allogeneic BMT, TBI oxidizes the lung GSH redox pool and Cy exacerbates this response by 5 days post-BMT. In contrast, liver GSH redox status is maintained under these experimental conditions. KGF treatment attenuates the Cy-induced decrease in lung GSH, decreases post-BMT lung lipid peroxidation, and improves liver GSH redox indices. KGF may have a therapeutic role to prevent or attenuate GSH depletion and ROS-mediated organ injury in BMT.


Asunto(s)
Trasplante de Médula Ósea , Factores de Crecimiento de Fibroblastos/farmacología , Glutatión/metabolismo , Hígado/metabolismo , Pulmón/metabolismo , Acondicionamiento Pretrasplante , Animales , Ciclofosfamida/farmacología , Femenino , Factor 7 de Crecimiento de Fibroblastos , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Oxidación-Reducción , Especies Reactivas de Oxígeno , Trasplante Homólogo , Irradiación Corporal Total
20.
Surgery ; 111(5): 591-4, 1992 May.
Artículo en Inglés | MEDLINE | ID: mdl-1598681

RESUMEN

Actinomycosis is an uncommon bacterial infection that has a characteristic chronic indolent course. Patients with this infection frequently undergo multiple surgical procedures before a correct diagnosis is made. Perianal actinomycosis should be suspected if a nontender perianal mass is found to contain thin purulent material and small yellow particles (sulfur granules). The diagnosis is confirmed by special stains and anaerobic cultures. Recognition of this infection is important because successful treatment requires combined surgical and antibiotic therapy. We report two patients, one with diabetes mellitus and one with human immunodeficiency virus III, who had recurrent perianal abscesses caused by Actinomyces and were treated successfully with surgical drainage and antimicrobial therapy.


Asunto(s)
Actinomicosis/complicaciones , Enfermedades del Ano/etiología , Diabetes Mellitus Tipo 1/complicaciones , Fístula/etiología , Infecciones por VIH/complicaciones , Seropositividad para VIH , Actinomyces/aislamiento & purificación , Actinomicosis/microbiología , Adulto , Enfermedades del Ano/patología , Enfermedades del Ano/cirugía , Fístula/patología , Fístula/cirugía , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda