Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bone ; 81: 562-572, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26318907

RESUMEN

Bone loss associated with microgravity exposure poses a significant barrier to long-duration spaceflight. Osteoprotegerin-Fc (OPG-Fc) is a receptor activator of nuclear factor kappa-B ligand (RANKL) inhibitor that causes sustained inhibition of bone resorption after a single subcutaneous injection. We tested the ability of OPG-Fc to preserve bone mass during 12 days of spaceflight (SF). 64-day-old female C57BL/6J mice (n=12/group) were injected subcutaneously with OPG-Fc (20mg/kg) or an inert vehicle (VEH), 24h prior to launch. Ground control (GC) mice (VEH or OPG-Fc) were maintained under environmental conditions that mimicked those in the space shuttle middeck. Age-matched baseline (BL) controls were sacrificed at launch. GC/VEH, but not SF/VEH mice, gained tibia BMD and trabecular volume fraction (BV/TV) during the mission (P<0.05 vs. BL). SF/VEH mice had lower BV/TV vs. GC/VEH mice, while SF/OPG-Fc mice had greater BV/TV than SF/VEH or GC/VEH. SF reduced femur elastic and maximum strength in VEH mice, with OPG-Fc increasing elastic strength in SF mice. Serum TRAP5b was elevated in SF/VEH mice vs. GC/VEH mice. Conversely, SF/OPG-Fc mice had lower TRAP5b levels, suggesting that OPG-Fc preserved bone during spaceflight via inhibition of osteoclast-mediated bone resorption. Decreased bone formation also contributed to the observed osteopenia, based on the reduced femur periosteal bone formation rate and serum osteocalcin level. Overall, these observations suggest that the beneficial effects of OPG-Fc during SF are primarily due to dramatic and sustained suppression of bone resorption. In growing mice, this effect appears to compensate for the SF-related inhibition of bone formation, while preventing any SF-related increase in bone resorption. We have demonstrated that the young mouse is an appropriate new model for SF-induced osteopenia, and that a single pre-flight treatment with OPG-Fc can effectively prevent the deleterious effects of SF on mouse bone.


Asunto(s)
Resorción Ósea/prevención & control , Fragmentos Fc de Inmunoglobulinas/farmacología , Osteoprotegerina/farmacología , Proteínas Recombinantes de Fusión/farmacología , Vuelo Espacial , Ingravidez/efectos adversos , Fosfatasa Alcalina/sangre , Animales , Biomarcadores/sangre , Fenómenos Biomecánicos , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/etiología , Resorción Ósea/fisiopatología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Osteocalcina/sangre , Ligando RANK/antagonistas & inhibidores
2.
Nat Rev Drug Discov ; 11(5): 401-19, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22543469

RESUMEN

Bone is a complex tissue that provides mechanical support for muscles and joints, protection for vital organs, a mineral reservoir that is essential for calcium homeostasis, and the environment and niches required for haematopoiesis. The regulation of bone mass in mammals is governed by a complex interplay between bone-forming cells termed osteoblasts and bone-resorbing cells termed osteoclasts, and is guided physiologically by a diverse set of hormones, cytokines and growth factors. The balance between these processes changes over time, causing an elevated risk of fractures with age. Osteoclasts may also be activated in the cancer setting, leading to bone pain, fracture, spinal cord compression and other significant morbidities. This Review chronicles the events that led to an increased understanding of bone resorption, the elucidation of the signalling pathway mediated by osteoprotegerin, receptor activator of NF-κB (RANK) and RANK ligand (RANKL) and its role in osteoclast biology, as well as the evolution of recombinant RANKL antagonists, which culminated in the development of the therapeutic RANKL-targeted antibody denosumab.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Osteoprotegerina/metabolismo , Ligando RANK/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Anticuerpos Monoclonales Humanizados , Resorción Ósea/fisiopatología , Huesos/metabolismo , Denosumab , Diseño de Fármacos , Humanos , Osteoclastos/fisiología , Ligando RANK/metabolismo , Transducción de Señal
3.
J Bone Miner Res ; 26(11): 2610-21, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21773994

RESUMEN

The physiological role of Dickkopf-1 (Dkk1) during postnatal bone growth in rodents and in adult rodents was examined utilizing an antibody to Dkk1 (Dkk1-Ab) that blocked Dkk1 binding to both low density lipoprotein receptor-related protein 6 (LRP6) and Kremen2, thereby preventing the Wnt inhibitory activity of Dkk1. Treatment of growing mice and rats with Dkk1-Ab resulted in a significant increase in bone mineral density because of increased bone formation. In contrast, treatment of adult ovariectomized rats did not appreciably impact bone, an effect that was associated with decreased Dkk1 expression in the serum and bone of older rats. Finally, we showed that Dkk1 plays a prominent role in adult bone by mediating fracture healing in adult rodents. These data suggest that, whereas Dkk1 significantly regulates bone formation in younger animals, its role in older animals is limited to pathologies that lead to the induction of Dkk1 expression in bone and/or serum, such as traumatic injury.


Asunto(s)
Envejecimiento/metabolismo , Huesos/lesiones , Huesos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Osteogénesis/fisiología , Envejecimiento/efectos de los fármacos , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/farmacología , Densidad Ósea/efectos de los fármacos , Enfermedades Óseas Metabólicas/sangre , Enfermedades Óseas Metabólicas/fisiopatología , Huesos/diagnóstico por imagen , Huesos/patología , Línea Celular , Estrógenos/deficiencia , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/patología , Curación de Fractura/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Masculino , Ratones , Osteogénesis/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Microtomografía por Rayos X
4.
J Bone Miner Res ; 26(5): 1012-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21542004

RESUMEN

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


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Densidad Ósea/efectos de los fármacos , Fracturas del Fémur/fisiopatología , Curación de Fractura/efectos de los fármacos , Glicoproteínas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Diáfisis/efectos de los fármacos , Diáfisis/patología , Diáfisis/fisiopatología , Modelos Animales de Enfermedad , Fémur/efectos de los fármacos , Fémur/patología , Fémur/fisiopatología , Peroné/efectos de los fármacos , Peroné/patología , Peroné/fisiopatología , Glicoproteínas/inmunología , Péptidos y Proteínas de Señalización Intercelular , Macaca fascicularis , Masculino , Tamaño de los Órganos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteotomía , Ratas , Ratas Sprague-Dawley
5.
Cell ; 142(4): 531-43, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20723755

RESUMEN

Muscle wasting and cachexia have long been postulated to be key determinants of cancer-related death, but there has been no direct experimental evidence to substantiate this hypothesis. Here, we show that in several cancer cachexia models, pharmacological blockade of ActRIIB pathway not only prevents further muscle wasting but also completely reverses prior loss of skeletal muscle and cancer-induced cardiac atrophy. This treatment dramatically prolongs survival, even of animals in which tumor growth is not inhibited and fat loss and production of proinflammatory cytokines are not reduced. ActRIIB pathway blockade abolished the activation of the ubiquitin-proteasome system and the induction of atrophy-specific ubiquitin ligases in muscles and also markedly stimulated muscle stem cell growth. These findings establish a crucial link between activation of the ActRIIB pathway and the development of cancer cachexia. Thus ActRIIB antagonism is a promising new approach for treating cancer cachexia, whose inhibition per se prolongs survival.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Caquexia/tratamiento farmacológico , Atrofia Muscular/tratamiento farmacológico , Miocardio/patología , Neoplasias/complicaciones , Receptores de Activinas Tipo II/genética , Activinas/metabolismo , Animales , Anorexia/tratamiento farmacológico , Anorexia/etiología , Atrofia/tratamiento farmacológico , Atrofia/etiología , Caquexia/etiología , Femenino , Humanos , Inhibinas/genética , Inhibinas/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/etiología , Mioblastos/patología , Trasplante de Neoplasias , Neoplasias/mortalidad , Transducción de Señal , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Bone Miner Res ; 25(5): 948-59, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20200929

RESUMEN

The development of bone-rebuilding anabolic agents for treating bone-related conditions has been a long-standing goal. Genetic studies in humans and mice have shown that the secreted protein sclerostin is a key negative regulator of bone formation. More recently, administration of sclerostin-neutralizing monoclonal antibodies in rodent studies has shown that pharmacologic inhibition of sclerostin results in increased bone formation, bone mass, and bone strength. To explore the effects of sclerostin inhibition in primates, we administered a humanized sclerostin-neutralizing monoclonal antibody (Scl-AbIV) to gonad-intact female cynomolgus monkeys. Two once-monthly subcutaneous injections of Scl-AbIV were administered at three dose levels (3, 10, and 30 mg/kg), with study termination at 2 months. Scl-AbIV treatment had clear anabolic effects, with marked dose-dependent increases in bone formation on trabecular, periosteal, endocortical, and intracortical surfaces. Bone densitometry showed that the increases in bone formation with Scl-AbIV treatment resulted in significant increases in bone mineral content (BMC) and/or bone mineral density (BMD) at several skeletal sites (ie, femoral neck, radial metaphysis, and tibial metaphysis). These increases, expressed as percent changes from baseline were 11 to 29 percentage points higher than those found in the vehicle-treated group. Additionally, significant increases in trabecular thickness and bone strength were found at the lumbar vertebrae in the highest-dose group. Taken together, the marked bone-building effects achieved in this short-term monkey study suggest that sclerostin inhibition represents a promising new therapeutic approach for medical conditions where increases in bone formation might be desirable, such as in fracture healing and osteoporosis.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Densidad Ósea/efectos de los fármacos , Proteínas Morfogenéticas Óseas/inmunología , Huesos/metabolismo , Marcadores Genéticos/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Huesos/patología , Femenino , Macaca fascicularis , Osteogénesis
7.
J Appl Physiol (1985) ; 106(2): 582-95, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074574

RESUMEN

Spaceflight results in a number of adaptations to skeletal muscle, including atrophy and shifts toward faster muscle fiber types. To identify changes in gene expression that may underlie these adaptations, we used both microarray expression analysis and real-time polymerase chain reaction to quantify shifts in mRNA levels in the gastrocnemius from mice flown on the 11-day, 19-h STS-108 shuttle flight and from normal gravity controls. Spaceflight data also were compared with the ground-based unloading model of hindlimb suspension, with one group of pure suspension and one of suspension followed by 3.5 h of reloading to mimic the time between landing and euthanization of the spaceflight mice. Analysis of microarray data revealed that 272 mRNAs were significantly altered by spaceflight, the majority of which displayed similar responses to hindlimb suspension, whereas reloading tended to counteract these responses. Several mRNAs altered by spaceflight were associated with muscle growth, including the phosphatidylinositol 3-kinase regulatory subunit p85alpha, insulin response substrate-1, the forkhead box O1 transcription factor, and MAFbx/atrogin1. Moreover, myostatin mRNA expression tended to increase, whereas mRNA levels of the myostatin inhibitor FSTL3 tended to decrease, in response to spaceflight. In addition, mRNA levels of the slow oxidative fiber-associated transcriptional coactivator peroxisome proliferator-associated receptor (PPAR)-gamma coactivator-1alpha and the transcription factor PPAR-alpha were significantly decreased in spaceflight gastrocnemius. Finally, spaceflight resulted in a significant decrease in levels of the microRNA miR-206. Together these data demonstrate that spaceflight induces significant changes in mRNA expression of genes associated with muscle growth and fiber type.


Asunto(s)
Regulación de la Expresión Génica , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Vuelo Espacial , Ingravidez , Adaptación Fisiológica/genética , Animales , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica/métodos , Suspensión Trasera , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/fisiopatología , Atrofia Muscular/fisiopatología , Miostatina/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/genética , Reacción en Cadena de la Polimerasa , Proteínas Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Serina-Treonina Quinasas TOR , Factores de Tiempo
8.
J Bone Miner Res ; 24(4): 578-88, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19049336

RESUMEN

The development of bone-rebuilding anabolic agents for potential use in the treatment of bone loss conditions, such as osteoporosis, has been a long-standing goal. Genetic studies in humans and mice have shown that the secreted protein sclerostin is a key negative regulator of bone formation, although the magnitude and extent of sclerostin's role in the control of bone formation in the aging skeleton is still unclear. To study this unexplored area of sclerostin biology and to assess the pharmacologic effects of sclerostin inhibition, we used a cell culture model of bone formation to identify a sclerostin neutralizing monoclonal antibody (Scl-AbII) for testing in an aged ovariectomized rat model of postmenopausal osteoporosis. Six-month-old female rats were ovariectomized and left untreated for 1 yr to allow for significant estrogen deficiency-induced bone loss, at which point Scl-AbII was administered for 5 wk. Scl-AbII treatment in these animals had robust anabolic effects, with marked increases in bone formation on trabecular, periosteal, endocortical, and intracortical surfaces. This not only resulted in complete reversal, at several skeletal sites, of the 1 yr of estrogen deficiency-induced bone loss, but also further increased bone mass and bone strength to levels greater than those found in non-ovariectomized control rats. Taken together, these preclinical results establish sclerostin's role as a pivotal negative regulator of bone formation in the aging skeleton and, furthermore, suggest that antibody-mediated inhibition of sclerostin represents a promising new therapeutic approach for the anabolic treatment of bone-related disorders, such as postmenopausal osteoporosis.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Proteínas Morfogenéticas Óseas/inmunología , Huesos/efectos de los fármacos , Marcadores Genéticos/inmunología , Osteogénesis/efectos de los fármacos , Osteoporosis Posmenopáusica/tratamiento farmacológico , Animales , Bioensayo , Fenómenos Biomecánicos , Densidad Ósea/efectos de los fármacos , Huesos/patología , Linaje de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fémur/efectos de los fármacos , Fémur/patología , Humanos , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Ratones , Pruebas de Neutralización , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteocalcina/sangre , Osteoporosis Posmenopáusica/sangre , Osteoporosis Posmenopáusica/patología , Osteoporosis Posmenopáusica/fisiopatología , Ovariectomía , Ratas , Ratas Sprague-Dawley , Tibia/efectos de los fármacos , Tibia/patología , Tomografía Computarizada por Rayos X
9.
J Bone Miner Res ; 23(6): 860-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18269310

RESUMEN

INTRODUCTION: Sclerosteosis is a rare high bone mass genetic disorder in humans caused by inactivating mutations in SOST, the gene encoding sclerostin. Based on these data, sclerostin has emerged as a key negative regulator of bone mass. We generated SOST knockout (KO) mice to gain a more detailed understanding of the effects of sclerostin deficiency on bone. MATERIALS AND METHODS: Gene targeting was used to inactivate SOST and generate a line of SOST KO mice. Radiography, densitometry, microCT, histomorphometry, and mechanical testing were used to characterize the impact of sclerostin deficiency on bone in male and female mice. Comparisons were made between same sex KO and wildtype (WT) mice. RESULTS: The results for male and female SOST KO mice were similar, with differences only in the magnitude of some effects. SOST KO mice had increased radiodensity throughout the skeleton, with general skeletal morphology being normal in appearance. DXA analysis of lumbar vertebrae and whole leg showed that there was a significant increase in BMD (>50%) at both sites. microCT analysis of femur showed that bone volume was significantly increased in both the trabecular and cortical compartments. Histomorphometry of trabecular bone revealed a significant increase in osteoblast surface and no significant change in osteoclast surface in SOST KO mice. The bone formation rate in SOST KO mice was significantly increased for trabecular bone (>9-fold) at the distal femur, as well as for the endocortical and periosteal surfaces of the femur midshaft. Mechanical testing of lumbar vertebrae and femur showed that bone strength was significantly increased at both sites in SOST KO mice. CONCLUSIONS: SOST KO mice have a high bone mass phenotype characterized by marked increases in BMD, bone volume, bone formation, and bone strength. These results show that sclerostin is a key negative regulator of a powerful, evolutionarily conserved bone formation pathway that acts on both trabecular and cortical bone.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Huesos/metabolismo , Eliminación de Gen , Osteogénesis , Proteínas Adaptadoras Transductoras de Señales , Animales , Biomarcadores/sangre , Densidad Ósea , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Huesos/diagnóstico por imagen , Calcio/sangre , Femenino , Marcadores Genéticos/genética , Glicoproteínas , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Noqueados , Fenotipo , Fosfatos/sangre , Estrés Mecánico , Tomografía Computarizada por Rayos X
10.
Bone ; 39(4): 754-66, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16730481

RESUMEN

Mutations affecting the activity of the Wnt co-receptors LRP5 and LRP6 that cause alterations in skeletal biology confirmed the involvement of Wnt signaling in bone formation. We evaluated the potential role of Dkk1, an inhibitor of LRP5/6 activity, in bone formation by examining the normal expression pattern of Dkk1 in normal young mice and by assessing the consequences of osteoblast overexpression of Dkk1 in transgenic mice. Endogenous Dkk1 expression was detected primarily in osteoblasts and osteocytes. Transgenic over-expression of Dkk1 using two different rat collagen 1A1 promoters resulted in distinct bone phenotypes. More widespread Dkk1 expression (driven by the Col1A1 3.6 kb promoter) yielded osteopenia with forelimb deformities and hairlessness, while expression restricted to osteoblasts (driven by the Col1A1 2.3 kb promoter) induced severe osteopenia without limb defects or alopecia. The decrease in bone mass in vivo resulted from a significant 49% reduction in osteoblast numbers and was reflected in a 45% reduction in serum osteocalcin concentration; an in vitro study revealed that Dkk1 caused a dose-dependent suppression of osteoblast matrix mineralization. These data indicate that Dkk1 may directly influence bone formation and suggest that osteopenia develops in mice over-expressing Dkk1 at least in part due to diminished bone formation resulting from reduced osteoblast numbers.


Asunto(s)
Enfermedades Óseas Metabólicas/fisiopatología , Huesos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas Wnt/fisiología , Células 3T3 , Animales , Densidad Ósea , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Huesos/patología , Huesos/fisiopatología , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas Relacionadas con Receptor de LDL/metabolismo , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/sangre , Osteogénesis/genética , Osteogénesis/fisiología , Embarazo , Ratas , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo
11.
Cancer Res ; 64(22): 8193-8, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15548684

RESUMEN

The progressive depletion of skeletal muscle is a hallmark of many types of advanced cancer and frequently is associated with debility, morbidity, and mortality. Muscle wasting is primarily mediated by the activation of the ubiquitin-proteasome system, which is responsible for degrading the bulk of intracellular proteins. E3 ubiquitin ligases control polyubiquitination, a rate-limiting step in the ubiquitin-proteasome system, but their direct involvement in muscle protein catabolism in cancer remains obscure. Here, we report the full-length cloning of E3alpha-II, a novel "N-end rule" ubiquitin ligase, and its functional involvement in cancer cachexia. E3alpha-II is highly enriched in skeletal muscle, and its expression is regulated by proinflammatory cytokines. In two different animal models of cancer cachexia, E3alpha-II was significantly induced at the onset and during the progression of muscle wasting. The E3alpha-II activation in skeletal muscle was accompanied by a sharp increase in protein ubiquitination, which could be blocked by arginine methylester, an E3alpha-selective inhibitor. Treatment of myotubes with tumor necrosis factor alpha or interleukin 6 elicited marked increases in E3alpha-II but not E3alpha-I expression and ubiquitin conjugation activity in parallel. E3alpha-II transfection markedly accelerated ubiquitin conjugation to endogenous cellular proteins in muscle cultures. These findings show that E3alpha-II plays an important role in muscle protein catabolism during cancer cachexia and suggest that E3alpha-II is a potential therapeutic target for muscle wasting.


Asunto(s)
Caquexia/metabolismo , Proteínas Musculares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Caquexia/enzimología , Clonación Molecular , ADN Complementario , Hidrólisis , Interleucina-6/fisiología , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Factor de Necrosis Tumoral alfa/fisiología , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/química
12.
Nature ; 423(6937): 337-42, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12748652

RESUMEN

Osteoclasts are specialized cells derived from the monocyte/macrophage haematopoietic lineage that develop and adhere to bone matrix, then secrete acid and lytic enzymes that degrade it in a specialized, extracellular compartment. Discovery of the RANK signalling pathway in the osteoclast has provided insight into the mechanisms of osteoclastogenesis and activation of bone resorption, and how hormonal signals impact bone structure and mass. Further study of this pathway is providing the molecular basis for developing therapeutics to treat osteoporosis and other diseases of bone loss.


Asunto(s)
Diferenciación Celular , Osteoclastos/citología , Osteoclastos/metabolismo , Animales , Resorción Ósea , Proteínas Portadoras/metabolismo , Glicoproteínas/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral , Transducción de Señal
13.
Am J Physiol Lung Cell Mol Physiol ; 285(3): L602-10, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12740217

RESUMEN

We reported an association between the ability of recombinant human keratinocyte growth factor (rHuKGF) to upregulate the expression of surfactant protein A (SP-A) and to downregulate pulmonary inflammation that occurs after allogeneic bone marrow transplantation (BMT). To establish a causal relationship, rHuKGF (5 mg/kg) was administered subcutaneously for three consecutive days before irradiation to SP-A-sufficient and -deficient [SP-A(+/+) and SP-A(-/-), respectively] mice given inflammation-inducing allogeneic spleen T cells at the time of BMT. In contrast with SP-A(+/+) mice, rHuKGF failed to suppress the high levels of TNF-alpha, IFN-gamma, and nitric oxide contained in bronchoalveolar lavage fluids collected on day 7 after BMT from SP-A(-/-) mice. Early post-BMT weight loss was attenuated by rHuKGF in both SP-A(+/+) and SP-A(-/-) recipients. In the absence of supportive respiratory care, however, SP-A deficiency eventually abolished the ability of rHuKGF to prevent weight loss and to improve survival monitored for 1 mo after allogeneic BMT. In further experiments, the addition of cyclophosphamide (which is known to cause severe injury to the alveolar epithelium in donor T cell-recipient mice) to the conditioning regimen prevented rHuKGF-induced upregulation of SP-A and suppression of lung inflammation in both SP-A(+/+) and SP-A(-/-) mice. We conclude that endogenous baseline SP-A levels and optimal upregulation of SP-A are required for the anti-inflammatory protective effects of KGF after allogeneic transplantation.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Factores de Crecimiento de Fibroblastos/farmacología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/metabolismo , Pulmón/metabolismo , Proteína A Asociada a Surfactante Pulmonar/genética , Animales , Trasplante de Médula Ósea/mortalidad , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Factor 7 de Crecimiento de Fibroblastos , Enfermedad Injerto contra Huésped/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Rendimiento Pulmonar/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteínas Recombinantes/farmacología , Linfocitos T/inmunología , Acondicionamiento Pretrasplante , Trasplante Homólogo , Regulación hacia Arriba/inmunología , Pérdida de Peso
14.
J Clin Invest ; 111(8): 1221-30, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12697741

RESUMEN

Studies in rodents have implicated various cytokines as paracrine mediators of increased osteoclastogenesis during estrogen deficiency, but increases in RANKL, the final effector of osteoclastogenesis, have not been demonstrated. Thus, we isolated bone marrow mononuclear cells expressing RANKL on their surfaces by two-color flow cytometry using FITC-conjugated osteoprotegerin-Fc (OPG-Fc-FITC) as a probe. The cells were characterized as preosteoblastic marrow stromal cells (MSCs), T lymphocytes, or B lymphocytes by using Ab's against bone alkaline phosphatase (BAP), CD3, and CD20, respectively, in 12 premenopausal women (Group A), 12 early postmenopausal women (Group B), and 12 age-matched, estrogen-treated postmenopausal women (Group C). Fluorescence intensity of OPG-Fc-FITC, an index of the surface concentration of RANKL per cell, was increased in Group B over Groups A and C by two- to threefold for MSCs, T cells, B cells, and total RANKL-expressing cells. Moreover, in the merged groups, RANKL expression per cell correlated directly with the bone resorption markers, serum C-terminal telopeptide of type I collagen and urine N-telopeptide of type I collagen, in all three cell types and inversely with serum 17beta-estradiol for total RANKL-expressing cells. The data suggest that upregulation of RANKL on bone marrow cells is an important determinant of increased bone resorption induced by estrogen deficiency.


Asunto(s)
Resorción Ósea/etiología , Proteínas Portadoras/fisiología , Glicoproteínas de Membrana/fisiología , Osteoporosis Posmenopáusica/etiología , Adulto , Proteínas Reguladoras de la Apoptosis , Femenino , Citometría de Flujo , Glicoproteínas/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Appl Physiol (1985) ; 94(5): 2085-94, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12514166

RESUMEN

There are several aspects of the spaceflight environment that may lead to changes in immunity: mission-related psychological stress, radiation, and changes in gravity. On December 5, 2001, the space shuttle Endeavor launched for a 12-day mission to examine these effects on C57BL/6 mice for the first time. On their return, assays were performed on the spleen, blood, and bone marrow. In response to flight, there were no significant differences in the general circulating leukocyte proportions. In contrast, there was an increase in splenic lymphocyte percentages, with a corresponding decrease in granulocytes. There was an overall shift in splenic lymphocytes away from T cells toward B cells, and a decrease in the CD4-to-CD8 ratios due to a decrease in T helpers. In contrast, there were proportional increases in bone marrow T cells, with decreases in B cells. Although the blast percentage and count were decreased in flight mice, the CD34(+) population was increased. The data were more consistent with a shift in bone marrow populations rather than a response to changes in the periphery. Many of the results are similar to those using other models. Clearly, spaceflight can influence immune parameters ranging from hematopoiesis to mature leukocyte mechanisms.


Asunto(s)
Inmunidad/fisiología , Vuelo Espacial , Animales , Médula Ósea/inmunología , Médula Ósea/fisiología , Femenino , Citometría de Flujo , Granulocitos/inmunología , Inmunidad Celular/inmunología , Inmunidad Celular/fisiología , Recuento de Leucocitos , Sistema Linfático/inmunología , Sistema Linfático/fisiología , Ratones , Ratones Endogámicos C57BL , Bazo/citología , Bazo/inmunología , Células Madre/fisiología
16.
J Appl Physiol (1985) ; 94(5): 2095-103, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12506046

RESUMEN

This portion of the study quantified the effects of a 12-day space shuttle mission (Space Transport System-108/UF-1) on body and lymphoid organ masses, activation marker expression, cytokine secretion, and erythrocyte and thrombocyte characteristics in C57BL/6 mice. Animals in flight (Flt group) had 10-12% lower body mass compared with ground controls housed either in animal enclosure modules or under standard vivarium conditions (P < 0.001) and the smallest thymus and spleen masses. Percentages of CD25(+) lymphocytes, CD3(+)/CD25(+) T cells, and NK1.1(+)/CD25(+) natural killer cells from Flt mice were higher compared with both controls (P < 0.05). In contrast, CD71 expression was depressed in the Flt and animal enclosure module control mice compared with vivarium control animals (P < 0.001). Secretion of interferon-gamma, IL-2, and IL-4, but not tumor necrosis factor-alpha and IL-5, by splenocytes from Flt mice was decreased relative to either one or both ground controls (P < 0.05). Flt mice also had high red blood cell and thrombocyte counts compared with both sets of controls; low red blood cell volume and distribution width, percentage of reticulocytes, and platelet volume were also noted (P < 0.05) and were consistent with dehydration. These data indicate that relatively short exposure to the spaceflight environment can induce profound changes that may become significant during long-term space missions.


Asunto(s)
Plaquetas/fisiología , Citocinas/biosíntesis , Eritrocitos/fisiología , Inmunidad/fisiología , Vuelo Espacial , Animales , Biomarcadores , Peso Corporal/fisiología , Supervivencia Celular/fisiología , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Recuento de Eritrocitos , Femenino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/fisiología , Recuento de Plaquetas , Bazo/citología
17.
Blood ; 100(2): 682-91, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12091365

RESUMEN

Thymus-dependent reconstitution of the peripheral T-cell compartment is critical for the successful outcome of bone marrow transplantation. However, graft-versus-host disease (GVHD) affects thymic stromal function and thus prevents normal T-cell maturation and selection. To determine whether cytoprotection of thymic epithelial cells (TECs) by keratinocyte growth factor (KGF) averts GVHD-related injury to the thymus, a nonirradiated murine parent-->F(1) transplantation model was investigated. Administration of KGF between days -3 and +3 of GVHD induction preserved normal thymic size, cellularity, and thymocyte phenotype when measured 2 weeks after transplantation and compared with saline-treated parent-->F(1) mice that received allogeneic transplants. Moreover, the characteristic GVHD-induced impairment in cell cycle progression of pro- and pre-T cells was prevented by KGF. However, the normal phenotypic and functional status of the thymus did not correlate with the higher number of GVHD-inducing mature donor T cells in thymi of KGF-treated mice. Importantly, extensive analysis of the different TEC populations within the thymic cortex and medulla revealed an almost normal stromal architecture and composition in GVHD mice treated with KGF. These observations are likely to reflect an indirect effect of KGF on thymopoiesis as KGF-receptor expression was demonstrated to be restricted to TECs. Thus, pharmacologic doses of KGF appear to exert a potent effect on TEC function, which in turn allows for normal T lymphopoiesis to occur during acute GVHD.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Enfermedad Injerto contra Huésped/prevención & control , Timo/citología , Animales , Antígeno B7-1/metabolismo , Ciclo Celular/efectos de los fármacos , Trasplante de Células/efectos adversos , Modelos Animales de Enfermedad , Células Epiteliales/citología , Factor 7 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/patología , Haplotipos , Ratones , Ratones Endogámicos C57BL , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/farmacología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Timo/efectos de los fármacos , Timo/inmunología
18.
Blood ; 99(12): 4592-600, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12036893

RESUMEN

Decreased thymopoietic capacity contributes to the severe and clinically significant immune deficiency seen after bone marrow transplantation (BMT). One mechanism for thymopoietic failure is damage to the interleukin 7 (IL-7)-producing thymic epithelial cells (TECs) by irradiation and chemotherapy, which can be partially treated by IL-7 administration. Pretreatment of BMT recipients with keratinocyte growth factor (KGF, or Fgf7), an epithelial cell-specific growth factor, protects mucosal, cutaneous, and pulmonary epithelial cells from cytotoxic therapy-induced damage in experimental murine models. Like other epithelial cells, TECs specifically express KGF receptors. Because KGF specifically protects KGF receptor-bearing epithelial cells and post-BMT immune deficiency is caused by loss of TECs, we hypothesized that KGF pretreatment would improve post-BMT thymic function. To test the hypothesis, BMT recipient mice were given KGF or placebo prior to congenic or allogeneic BMT. Administration of KGF before murine BMT significantly increased the capacity of the thymus to generate donor-derived thymocytes. KGF pretreatment also normalized the proportion of thymic subpopulations, increased the number of naive T cells in the periphery, and improved the response to neoantigen immunization. KGF treatment caused increased production of intrathymic IL-7, and the thymopoietic effects of KGF required an intact IL-7 signaling pathway. These results demonstrate that KGF may have immunomodulatory effects by a unique mechanism of protection of TECs. Furthermore, thymic injury and prolonged posttransplantation immune deficiency in BMT recipients can be prevented by KGF administration.


Asunto(s)
Trasplante de Médula Ósea/métodos , Células Epiteliales/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Sustancias Protectoras/farmacología , Timo/efectos de los fármacos , Animales , Células Sanguíneas/efectos de los fármacos , Trasplante de Médula Ósea/normas , Células Epiteliales/citología , Factor 7 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Supervivencia de Injerto/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/crecimiento & desarrollo , Ratones , Ratones Endogámicos BALB C , Sustancias Protectoras/administración & dosificación , Linfocitos T/efectos de los fármacos , Timo/citología
19.
Am J Physiol Gastrointest Liver Physiol ; 282(4): G690-701, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11897629

RESUMEN

There is an acute need for effective therapy for inflammatory bowel disease (IBD), particularly at the level of repair of the damaged epithelium. We evaluated the efficacy of recombinant human keratinocyte growth factor (rHuKGF) in both the dextran sodium sulfate (DSS) and the CD4(+)CD45RB(Hi) T cell transfer models of IBD. Disease was induced either by the ad libitum administration to normal mice of 4% DSS in the drinking water or by the injection of 4 x 10(5) CD4(+)CD45RB(Hi) T cells into immunodeficient scid/scid mice. rHuKGF was administered by subcutaneous injection at doses of 1.0 or 3.0 mg/kg in both preventative and therapeutic regimens during both studies. rHuKGF significantly improved survival and body weight loss in the DSS model in both preventative and therapeutic dosing regimens. It also improved diarrhea, hematochezia, and hematological parameters, as well as large intestine histopathology. In the T cell transfer model, rHuKGF improved body weight loss, diarrhea, and levels of serum amyloid A, as well as large intestine histopathology. In both models of IBD, the colonic levels of intestinal trefoil factor (ITF) were elevated by the disease state and further elevated by treatment with rHuKGF. These data suggest that rHuKGF may prove useful in the clinical management of IBD and its effects are likely mediated by its ability to locally increase the levels of ITF.


Asunto(s)
Linfocitos T CD4-Positivos/trasplante , Sulfato de Dextran , Factores de Crecimiento de Fibroblastos/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Antígenos Comunes de Leucocito/análisis , Animales , Linfocitos T CD4-Positivos/inmunología , Diarrea , Modelos Animales de Enfermedad , Femenino , Factor 7 de Crecimiento de Fibroblastos , Humanos , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/fisiopatología , Mucosa Intestinal/patología , Recuento de Leucocitos , Masculino , Ratones , Ratones SCID , Neutrófilos , Proteínas Recombinantes/uso terapéutico , Proteína Amiloide A Sérica/análisis , Pérdida de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...