Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Am J Physiol Endocrinol Metab ; 304(3): E282-93, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23211515

RESUMEN

Glucocorticoids are extremely effective anti-inflammatory therapies, but their clinical use is limited due to severe side effects, including osteoporosis, muscle wasting, fat redistribution, and skin thinning. Here we use heavy water labeling and mass spectrometry to measure fluxes through metabolic pathways impacted by glucocorticoids. We combine these methods with measurements of body composition in corticotropin-releasing hormone (CRH)-transgenic (Tg)(+) mice that have chronically elevated, endogenously produced corticosterone and a phenotype that closely mimics Cushing's disease in humans. CRH-Tg(+) mice had increased adipose mass, adipose triglyceride synthesis, and greatly increased triglyceride/fatty acid cycling in subcutaneous and abdominal fat depots and increased de novo lipogenesis in the abdominal depot. In bone, CRH-Tg(+) mice had decreased bone mass, absolute collagen synthesis rates, and collagen breakdown rate. In skin, CRH-Tg(+) mice had decreased skin thickness and absolute collagen synthesis rates but no decrease in the collagen breakdown rate. In muscle, CRH-Tg(+) mice had decreased muscle mass and absolute protein synthesis but no decrease in the protein breakdown rate. We conclude that chronic exposure to endogenous glucocorticoid excess in mice is associated with ongoing decreases in bone collagen, skin collagen, and muscle protein synthesis without compensatory reduction (coupling) of breakdown rates in skin and muscle. Both of these actions contribute to reduced protein pool sizes. We also conclude that increased cycling between triglycerides and free fatty acids occurs in both abdominal and subcutaneous fat depots in CRH-Tg(+) mice. CRH-Tg mice have both increased lipolysis and increased triglyceride synthesis in adipose tissue.


Asunto(s)
Tejido Adiposo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/metabolismo , Triglicéridos/metabolismo , Adiposidad , Animales , Hormona Liberadora de Corticotropina/genética , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
2.
Am J Physiol Endocrinol Metab ; 303(11): E1354-62, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23047986

RESUMEN

Loss of skeletal weight bearing or skeletal unloading as occurs during spaceflight inhibits bone formation and stimulates bone resorption. These are associated with a decline in the osteoblast (Ob.S/BS) and an increase in the osteoclast (Oc.S/BS) bone surfaces. To determine the temporal relationship between changes in the bone cells and their marrow precursor pools during sustained unloading, and whether genetic background influences these relationships, we used the hindlimb unloading model to induce bone loss in two strains of mice known to respond to load and having significantly different cancellous bone volumes (C57BL/6 and DBA/2 male mice). Skeletal unloading caused a progressive decline in bone volume that was accompanied by strain-specific changes in Ob.S/BS and Oc.S/BS. These were associated with a sustained reduction in the osteoprogenitor population and a dramatic but transient increase in the osteoclast precursor pool size in both strains. The results reveal that bone adaptation to skeletal unloading involves similar rapid changes in the osteoblast and osteoclast progenitor populations in both strains of mice but striking differences in Oc.S/BS dynamics, BFR, and cancellous bone structure. These strain-specific differences suggest that genetics plays an important role in determining the osteoblast and osteoclast populations on the bone surface and the dynamics of bone loss in response to skeletal unloading.


Asunto(s)
Remodelación Ósea/fisiología , Resorción Ósea/patología , Huesos/citología , Suspensión Trasera/fisiología , Osteoblastos/fisiología , Osteoclastos/fisiología , Animales , Células de la Médula Ósea/citología , Resorción Ósea/fisiopatología , Huesos/patología , Diferenciación Celular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Vuelo Espacial , Simulación del Espacio , Especificidad de la Especie , Células Madre/citología
3.
Proc Natl Acad Sci U S A ; 105(4): 1209-14, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18212126

RESUMEN

Osteoblasts are essential for maintaining bone mass, avoiding osteoporosis, and repairing injured bone. Activation of osteoblast G protein-coupled receptors (GPCRs), such as the parathyroid hormone receptor, can increase bone mass; however, the anabolic mechanisms are poorly understood. Here we use "Rs1," an engineered GPCR with constitutive G(s) signaling, to evaluate the temporal and skeletal effects of G(s) signaling in murine osteoblasts. In vivo, Rs1 expression induces a dramatic anabolic skeletal response, with midfemur girth increasing 1,200% and femur mass increasing 380% in 9-week-old mice. Bone volume, cellularity, areal bone mineral density, osteoblast gene markers, and serum bone turnover markers were also elevated. No such phenotype developed when Rs1 was expressed after the first 4 weeks of postnatal life, indicating an exquisite temporal sensitivity of osteoblasts to Rs1 expression. This pathway may represent an important determinant of bone mass and may open future avenues for enhancing bone repair and treating metabolic bone diseases.


Asunto(s)
Densidad Ósea/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Osteoblastos/química , Osteoblastos/metabolismo , Ingeniería de Proteínas , Receptores de Serotonina 5-HT4/biosíntesis , Receptores de Serotonina 5-HT4/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Humanos , Ligandos , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Ingeniería de Proteínas/métodos , Agonistas del Receptor de Serotonina 5-HT4 , Transducción de Señal/genética , Transducción de Señal/fisiología
4.
J Bone Miner Res ; 22(9): 1329-37, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17539737

RESUMEN

UNLABELLED: We showed that the IGF-IR-null mutation in mature osteoblasts leads to less bone and decreased periosteal bone formation and impaired the stimulatory effects of PTH on osteoprogenitor cell proliferation and differentiation. INTRODUCTION: This study was carried out to examine the role of IGF-I signaling in mediating the actions of PTH on bone. MATERIALS AND METHODS: Three-month-old mice with an osteoblast-specific IGF-I receptor null mutation (IGF-IR OBKO) and their normal littermates were treated with vehicle or PTH (80 microg/kg body weight/d for 2 wk). Structural measurements of the proximal and midshaft of the tibia were made by microCT. Trabecular and cortical bone formation was measured by bone histomorphometry. Bone marrow stromal cells (BMSCs) were obtained to assess the effects of PTH on osteoprogenitor number and differentiation. RESULTS: The fat-free weight of bone normalized to body weight (FFW/BW), bone volume (BV/TV), and cortical thickness (C.Th) in both proximal tibia and shaft were all less in the IGF-IR OBKO mice compared with controls. PTH decreased FFW/BW of the proximal tibia more substantially in controls than in IGF-IR OBKO mice. The increase in C.Th after PTH in the proximal tibia was comparable in both control and IGF-IR OBKO mice. Although trabecular and periosteal bone formation was markedly lower in the IGF-IR OBKO mice than in the control mice, endosteal bone formation was comparable in control and IGF-IR OBKO mice. PTH stimulated endosteal bone formation only in the control animals. Compared with BMSCs from control mice, BMSCs from IGF-IR OBKO mice showed equal alkaline phosphatase (ALP)(+) colonies on day 14, but fewer mineralized nodules on day 28. Administration of PTH increased the number of ALP(+) colonies and mineralized nodules on days 14 and 28 in BMSCs from control mice, but not in BMSCs from IGF-IR OBKO mice. CONCLUSIONS: Our results indicate that the IGF-IR null mutation in mature osteoblasts leads to less bone and decreased bone formation, in part because of the requirement for the IGF-IR in mature osteoblasts to enable PTH to stimulate osteoprogenitor cell proliferation and differentiation.


Asunto(s)
Huesos/fisiología , Hormona Paratiroidea/fisiología , Receptor IGF Tipo 1/fisiología , Animales , Secuencia de Bases , Biomarcadores/metabolismo , Peso Corporal , Proliferación Celular , Células Cultivadas , Cartilla de ADN , Ratones , Ratones Noqueados , Mutación , Tamaño de los Órganos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
5.
J Appl Physiol (1985) ; 103(1): 125-31, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17412794

RESUMEN

IGF-I stimulates osteoblast proliferation, bone formation, and increases bone volume in normal weight-bearing animals. During skeletal unloading or loss of weight bearing, bone becomes unresponsive to the anabolic effects of insulin-like growth factor I (IGF-I). To determine whether skeletal reloading after a period of unloading increases bone responsiveness to IGF-I, we examined bone structure and formation in response to IGF-I under different loading conditions. Twelve-week-old rats were divided into six groups: loaded (4 wk), unloaded (4 wk), and unloaded/reloaded (2/2 wk), and treated with IGF-I (2.5 mg x kg(-1) x day(-1)) or vehicle during the final 2 wk. Cortical bone formation rate (BFR), cancellous bone volume and architecture in the secondary spongiosa (tibia and vertebrae), and total volume and calcified volume in the primary spongiosa (tibia) were assessed. Periosteal BFR decreased during unloading, remained low during reloading in the vehicle-treated group, but was dramatically increased in IGF-I-treated animals. Cancellous bone volume decreased with unloading and increased with reloading, but the effect was exaggerated in the tibia of IGF-I-treated animals. Total and calcified volumes in the primary spongiosa decreased during unloading in the vehicle-treated animals. IGF-I treatment prevented the loss in volume. These data show that reloading after a period of skeletal unloading increases bone responsiveness to IGF-I, and they suggest that IGF-I may be of therapeutic use in patients who have lost bone as a consequence of prolonged skeletal disuse.


Asunto(s)
Regeneración Ósea , Resorción Ósea/metabolismo , Huesos/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Densidad Ósea , Regeneración Ósea/efectos de los fármacos , Resorción Ósea/patología , Resorción Ósea/fisiopatología , Huesos/efectos de los fármacos , Huesos/patología , Huesos/fisiopatología , Modelos Animales de Enfermedad , Peroné/metabolismo , Peroné/fisiopatología , Suspensión Trasera , Húmero/metabolismo , Húmero/fisiopatología , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Columna Vertebral/metabolismo , Columna Vertebral/fisiopatología , Tibia/metabolismo , Tibia/fisiopatología , Factores de Tiempo , Tomografía Computarizada por Rayos X , Soporte de Peso
6.
J Bone Miner Res ; 25(3): 584-93, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20200944

RESUMEN

Age-dependent changes in skeletal growth are important for regulating skeletal expansion and determining peak bone mass. However, how G protein-coupled receptors (GPCRs) regulate these changes is poorly understood. Previously, we described a mouse model expressing Rs1, an engineered receptor with high basal G(s) activity. Rs1 expression in osteoblasts induced a dramatic age-dependent increase in trabecular bone with features resembling fibrous dysplasia. To further investigate how activation of the G(s)-GPCR pathway affects bone formation at different ages, we used the tetracycline-inducible system in the ColI(2.3)(+)/Rs1(+) mouse model to control the timing of Rs1 expression. We found that the Rs1 phenotype developed rapidly between postnatal days 4 and 6, that delayed Rs1 expression resulted in attenuation of the Rs1 phenotype, and that the Rs1-induced bone growth and deformities were markedly reversed when Rs1 expression was suppressed in adult mice. These findings suggest a distinct window of increased osteoblast responsiveness to G(s) signaling during the early postnatal period. In addition, adult bones encode information about their normal shape and structure independently from mechanisms regulating bone expansion. Finally, our model provides a powerful tool for investigating the effects of continuous G(s)-GPCR signaling on dynamic bone growth and remodeling.


Asunto(s)
Osteoblastos/metabolismo , Osteogénesis/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Factores de Edad , Animales , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA