Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 27(9): 3505-13, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23704087

RESUMEN

The chemokine CXCL12 and its receptor CXCR4 play a key role in regulation of hematopoietic stem cells and cell migratory function during morphogenesis. Osteoblasts express both the ligand and the receptor, but little is known about the role of CXCL12-CXCR4 signaling in maintaining skeletal homeostasis. Using Cre-Lox technology to delete CXCR4 in mature osteoblasts in mice, we show here a significant decrease in bone mass and alterations in cancellous bone structure. CXCR4 gene ablation increased the number of colony-forming units (CFU), CFU-positive for alkaline phosphatase (CFU-AP(+)), and mineralizing nodules in bone marrow stromal cell (BMSC) cultures. The adipocyte precursor population decreased in BMSCs harvested from the KO animals. The nonadherent population of BMSCs harvested from the long bone diaphysis of KO animals formed more osteoclasts, a finding that was associated with increased circulatory levels of pyridinoline, a marker of bone resorption. Our data show that osteoblast-specific CXCR4 deletion has profound effects on the mesenchymal stem cell pool and allocation to the osteoblastic and adipocytic cell lineages. They also show that CXCL12/CXCR4 signaling in the mature osteoblast can feedback to regulate the osteoclast precursor pool size and play a multifunctional role in regulating bone formation and resorption.


Asunto(s)
Quimiocina CXCL12/metabolismo , Células Madre Mesenquimatosas/citología , Osteoclastos/citología , Receptores CXCR4/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/farmacología , Genotipo , Inmunohistoquímica , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Noqueados , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Receptores CXCR4/genética , Microtomografía por Rayos X
2.
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
3.
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
4.
Calcif Tissue Int ; 91(1): 50-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22644321

RESUMEN

Sclerostin functions as an antagonist to Wnt signaling and inhibits bone-forming activity. We studied the effects of skeletal unloading and treatment with sclerostin antibody (Scl-Ab) on mesenchymal stem cell, osteoprogenitor and osteoclast precursor pools, and their relationship to bone formation and resorption. Male C57BL/6 mice (5-months-old) were hind limb unloaded for 1 week or allowed normal ambulation and treated with Scl-Ab (25 mg/kg, s.c. injections on days 1 and 4) or placebo. Unloading decreased the serum concentration of bone formation marker P1NP (-35 %), number of colony-forming units (CFU) (-38 %), alkaline phosphatase-positive CFUs (CFU-AP+) (-51 %), and calcified nodules (-35 %); and resulted in a fourfold increase in the number of osteoclast precursors. The effects of Scl-Ab treatment on unloaded and normally loaded mice were nearly identical; Scl-Ab increased serum P1NP and the number of CFU, CFU-AP+, and calcified nodules in ex vivo cultures; and increased osteoblast and bone mineralizing surfaces in vivo. Although the marrow-derived osteoclast precursor population increased with Scl-Ab, the bone osteoclast surface did not change, and the serum concentration of osteoclast activity marker TRACP5b decreased. Our data suggest that short-term Scl-Ab treatment can prevent the decrease in osteoprogenitor population associated with skeletal unloading and increase osteoblast surface and bone mineralizing surface in unloaded animals. The anabolic effects of Scl-Ab treatment on bone are preserved during skeletal unloading. These findings suggest that Scl-Ab treatment can both increase bone formation and decrease bone resorption, and provide a new means for prevention and treatment of disuse osteoporosis.


Asunto(s)
Anticuerpos/metabolismo , Médula Ósea/metabolismo , Glicoproteínas/inmunología , Fosfatasa Ácida/genética , Fosfatasa Ácida/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Resorción Ósea , Glicoproteínas/genética , Glicoproteínas/metabolismo , Suspensión Trasera , Péptidos y Proteínas de Señalización Intercelular , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Células Madre/citología , Células Madre/metabolismo , Fosfatasa Ácida Tartratorresistente
5.
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
6.
Int J Spine Surg ; 15(5): 921-928, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34649952

RESUMEN

BACKGROUND: Ascorbic acid is involved in collagen biosynthesis and upregulates alkaline phosphatase, potentially alleviating cell senescence and stimulating mesenchymal stem cell proliferation and differentiation into osteoblasts. We hypothesized locally delivered ascorbic acid and ß-glycerophosphate act as a bone graft extender to increase the volume of new bone formed in a murine model of posterior lumbar fusion. METHODS: Collagen sponges were used as delivery vehicles. Sponges were prepared with primary media alone or with the addition of ascorbic acid and ß-glycerophosphate. Fresh morselized bone graft from 12 donor mice was used. Twenty-four healthy male C57BL/6 mice underwent an uninstrumented posterior L3-L5 lumbar fusion. One control group received morselized bone only. A second "sponge control" group received morselized bone with the control collagen sponge. The third group received morselized bone and a collagen sponge with ascorbic acid and ß-glycerophosphate. Three months postoperatively, the lumbar spine underwent high-resolution micro-computed tomography for analysis of bone formation, density, and bridging fusion. RESULTS: Animals receiving ascorbic acid and ß-glycerophosphate had a statistically significant increase in corrected bone volume compared with control and sponge groups, with a 56.3% and 25.4% increase, respectively. Mineralized bone fraction was statistically significantly decreased for animals in the ascorbic acid group compared with control and sponge groups. There was no significant difference in fusion rate between test groups. CONCLUSIONS: Locally delivered ascorbic acid and ß-glycerophosphate in a murine model of posterior spinal fusion yielded statistically significant increases in new bone formation in the lumbar spine but statistically significant decreases in mineralized bone fraction. Differences in fusion rate were not statistically significant. CLINICAL RELEVANCE: This study provides early data suggesting that delivery of ascorbic acid to a spinal fusion site may be beneficial but does not yet establish an indication for clinical use. Further studies are needed to determine optimal dose and delivery of ascorbic acid.

7.
J Nutr ; 140(10): 1781-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20739449

RESUMEN

Bone is progressively lost with advancing age. Therapies are limited and the only effective proanabolic regimen presently available to restore bone is intermittent treatment with teriparatide (parathyroid hormone 1-34). Recent evidence suggests that dietary supplementation with dried plum (DP) can prevent bone loss due to estrogen deficiency. To determine whether dietary DP supplementation can prevent the loss of bone with aging and whether bone that has already been lost can be restored, adult (6 mo) and old (18 mo) male mice were fed a normal diet or isoenergetic, isonitrogenous diets supplemented with DP (0, 15, and 25% DP by weight) for 6 mo. MicroCT analysis and bone histomorphometry were used to assess bone volume, structure, and metabolic activity before, during, and after dietary supplementation. Mice fed the 0% DP diet (control diet) lost bone, whereas both adult and old mice fed the 25% DP-supplemented diet gained bone. Adult but not old mice fed the 15% diet also gained bone. Cancellous bone volume in mice receiving 25% DP exceeded baseline levels by 40-50%. Trabecular structure varied with diet and age and responses in old mice were generally blunted. Trabecular, but not cortical, mineral density varied with age and measures of bone anabolic activity were lower in aged mice. Our findings suggest that DP contains proanabolic factors that can dramatically increase bone volume and restore bone that has already been lost due to aging. In turn, DP may provide effective prophylactic and therapeutic agents for the treatment of osteoporosis.


Asunto(s)
Envejecimiento , Dieta , Alimentos en Conserva , Frutas , Osteoporosis/prevención & control , Prunus , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Densidad Ósea , Huesos/metabolismo , Huesos/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Fitoterapia
8.
JOR Spine ; 3(4): e1113, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33392451

RESUMEN

Spinal cord injury (SCI) is accompanied by rapid loss of bone and increased risk of low impact fractures. Current pharmacological treatment approaches have proven to be relatively ineffective in preventing or treating bone loss after SCI. Dietary supplementation with dried plum (DP) has been shown to have dramatic effects on bone in various other disease models. In this study, we tested the efficacy of DP in preventing bone loss after SCI and restoring bone that has already been lost in response to SCI. Male C57BL/6J mice (3-month-old) underwent SCI and were fed a diet containing 25% DP by weight or a control diet for up to 4 weeks to assess whether DP can prevent bone loss. To determine whether DP could restore bone already lost due to SCI, mice were put on a control diet for 2 weeks (to allow bone loss) and then shifted to a DP supplemented diet for an additional 2 weeks. The skeletal responses to SCI and dietary supplementation with DP were assessed using microCT analysis, bone histomorphometry and strength testing. Dietary supplementation with DP completely prevented the loss of bone and bone strength induced by SCI in acutely injured mice. DP also could restore a fraction of the bone lost and attenuate the loss of bone strength after SCI. These results suggest that dietary supplementation with DP or factors derived from DP may prove to be an effective treatment for the loss of bone in patients with SCI.

9.
Sci Rep ; 10(1): 6484, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32300161

RESUMEN

Spaceflight is a unique environment that includes at least two factors which can negatively impact skeletal health: microgravity and ionizing radiation. We have previously shown that a diet supplemented with dried plum powder (DP) prevented radiation-induced bone loss in mice. In this study, we investigated the capacity of the DP diet to prevent bone loss in mice following exposure to simulated spaceflight, combining microgravity (by hindlimb unloading) and radiation exposure. The DP diet was effective at preventing most decrements in bone micro-architectural and mechanical properties due to hindlimb unloading alone and simulated spaceflight. Furthermore, we show that the DP diet can protect osteoprogenitors from impairments resulting from simulated microgravity. Based on our findings, a dietary supplementation with DP could be an effective countermeasure against the skeletal deficits observed in astronauts during spaceflight.


Asunto(s)
Enfermedades Óseas Metabólicas/prevención & control , Radiación Cósmica/efectos adversos , Suspensión Trasera/efectos adversos , Prunus domestica , Vuelo Espacial , Animales , Densidad Ósea/fisiología , Densidad Ósea/efectos de la radiación , Enfermedades Óseas Metabólicas/diagnóstico , Enfermedades Óseas Metabólicas/etiología , Enfermedades Óseas Metabólicas/fisiopatología , Modelos Animales de Enfermedad , Alimentos en Conserva , Suspensión Trasera/fisiología , Humanos , Masculino , Ratones , Esqueleto/diagnóstico por imagen , Esqueleto/fisiopatología , Esqueleto/efectos de la radiación , Microtomografía por Rayos X
10.
J Bone Miner Res ; 22(8): 1271-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17488198

RESUMEN

UNLABELLED: IGF-I plays an important anabolic role in stimulating bone formation and maintaining bone mass. We show that the pro-proliferative, anti-apoptotic, and functional responses to IGF-I in bone and BMSCs decrease with aging. These changes are associated with impaired receptor activation and signal transduction through the MAPK and PI3K pathways. INTRODUCTION: IGF-I is a potent anabolic agent having effects across diverse tissues and cell types. With aging, bone becomes resistant to the anabolic actions of IGF-I. To examine the effects of aging on bone responsiveness to IGF-I, we measured the pro-proliferative, anti-apoptotic, and functional responses of bone and bone marrow stromal cells (BMSCs) to IGF-I and evaluated IGF-I signal transduction in young, adult, and old mice. MATERIALS AND METHODS: Male C57BL/6 mice 6 wk (young), 6 mo (adult), and 24 mo (old) were treated with IGF-I for 2 wk using osmotic minipumps, and osteoblast proliferation (BrdU labeling) in vivo, and osteoprogenitor number (BMSC culture and calcium nodule formation) were measured. Proliferation, apoptosis, and expression of key osteoblast factors (alkaline phosphatase, collagen, osteocalcin, RANKL, osteoprotegerin (OPG), macrophage-colony stimulating factor [M-CSF]) and IGF-I signaling elements and their activation in IGF-I-treated cells were studied using QRT-PCR and Western blot analysis. Data were analyzed using ANOVA. RESULTS: Aging decreased the basal and IGF-I-stimulated number of BrdU-labeled osteoblasts and reduced the ability of IGF-I to stimulate osteoprogenitor formation (calcium nodule number) by 50%. The pro-proliferative and anti-apoptotic actions of IGF-I were blunted in cells from old animals. These changes were accompanied by age-related alterations in the ability of IGF-I to regulate alkaline phosphatase, collagen, osteocalcin, RANKL, OPG, and M-CSF expression. IGF-I binding was normal, but IGF-I receptor mRNA and protein expression was increased in aged animals by 2- and 10-fold, respectively. The age-related changes in proliferation, apoptosis, and function were accompanied by loss of IGF-I-induced signaling at the receptor level and at key regulatory sites along the MAPK (ERK1/2) and PI3K (AKT) pathways. CONCLUSIONS: Our data show that aging is accompanied by loss of bone and BMSC/osteoblast responsiveness to IGF-I and that these changes are associated with resistance to IGF-I signaling that involve receptor activation and downstream signaling events.


Asunto(s)
Envejecimiento/fisiología , Huesos/efectos de los fármacos , Huesos/enzimología , Factor I del Crecimiento Similar a la Insulina/farmacología , Receptor IGF Tipo 1/metabolismo , Fosfatasa Alcalina/genética , Animales , Células Cultivadas , Colágeno/genética , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Ratones , Osteocalcina/genética , Osteoclastos/efectos de los fármacos , Osteoclastos/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Receptor IGF Tipo 1/genética
11.
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
12.
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
13.
J Bone Miner Res ; 21(9): 1350-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16939393

RESUMEN

UNLABELLED: We showed that IGF-I deficiency impaired osteoclastogenesis directly and/or indirectly by altering the interaction between stromal/osteoblastic cells and osteoclast precursors, reducing RANKL and M-CSF production. These changes lead to impaired bone resorption, resulting in high BV/TV in IGF-I null mice. INTRODUCTION: Although IGF-I has been clearly identified as an important growth factor in regulating osteoblast function, information regarding its role in osteoclastogenesis is limited. Our study was designed to analyze the role of IGF-I in modulating osteoclastogenesis using IGF-I knockout mice (IGF-I(-/-)). MATERIALS AND METHODS: Trabecular bone volume (BV/TV), osteoclast number, and morphology of IGF-I(-/-) or wildtype mice (IGF-I(+/+)) were evaluated in vivo by histological analysis. Osteoclast precursors from these mice were cultured in the presence of RANKL and macrophage-colony stimulating factor (M-CSF) or co-cultured with stromal/osteoblastic cells from either genotype. Osteoclast formation was assessed by measuring the number of multinucleated TRACP+ cells and pit formation. The mRNA levels of osteoclast regulation markers were determined by quantitative RT-PCR. RESULTS: In vivo, IGF-I(-/-) mice have higher BV/TV and fewer (76% of IGF-I(+/+)) and smaller osteoclasts with fewer nuclei. In vitro, in the presence of RANKL and M-CSF, osteoclast number (55% of IGF-I(+/+)) and resorptive area (30% of IGF-I(+/+)) in osteoclast precursor cultures from IGF-I(-/-) mice were significantly fewer and smaller than that from the IGF-I(+/+) mice. IGF-I (10 ng/ml) increased the size, number (2.6-fold), and function (resorptive area, 2.7-fold) of osteoclasts in cultures from IGF-I(+/+) mice, with weaker stimulation in cultures from IGF-I(-/-) mice. In co-cultures of IGF-I(-/-) osteoblasts with IGF-I(+/+) osteoclast precursors, or IGF-I(+/+) osteoblasts with IGF-I(-/-) osteoclast precursors, the number of osteoclasts formed was only 11% and 48%, respectively, of that from co-cultures of IGF-I(+/+) osteoblasts and IGF-I(+/+) osteoclast precursors. In the long bones from IGF-I(-/-) mice, mRNA levels of RANKL, RANK, M-CSF, and c-fms were 55%, 33%, 60%, and 35% of that from IGF-I(+/+) mice, respectively. CONCLUSIONS: Our results indicate that IGF-I regulates osteoclastogenesis by promoting their differentiation. IGF-I is required for maintaining the normal interaction between the osteoblast and osteoclast to support osteoclastogenesis through its regulation of RANKL and RANK expression.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/fisiología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Resorción Ósea/genética , Huesos/fisiología , Proteínas Portadoras/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Glicoproteínas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Técnicas In Vitro , Factor I del Crecimiento Similar a la Insulina/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Osteoclastos/fisiología , Osteogénesis/genética , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores de Calcitonina/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología
14.
Endocrinology ; 147(10): 4753-61, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16857753

RESUMEN

Although IGF-I has been identified as an important growth factor for the skeleton, the role of IGF-I on embryonic bone development remains unknown. Here we show that, in IGF-I-deficient (IGF-I(-/-)) mice, skeletal malformations, including short-limbed dwarfism, were evident at days post coitus (dpc) 14.5 to 18.5, accompanied by delays of mineralization in the spinal column, sternum, and fore paws. Reduced chondrocyte proliferation and increased chondrocyte apoptosis were identified in both the spinal ossification center and the growth plate of long bones. Abnormal chondrocyte differentiation and delayed initiation of mineralization was characterized by small size and fewer numbers of type X collagen expressing hypertrophic chondrocytes and lower osteocalcin expression. The Indian hedgehog-PTHrP feedback loop was altered; expression of Indian hedgehog was reduced in IGF-I(-/-) mice in long bones and in the spine, whereas expression of PTHrP was increased. Our results indicate that IGF-I plays an important role in skeletal development by promoting chondrocyte proliferation and maturation while inhibiting apoptosis to form bones of appropriate size and strength.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/embriología , Factor I del Crecimiento Similar a la Insulina/fisiología , Animales , Huesos/citología , Calcificación Fisiológica/genética , Calcificación Fisiológica/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Condrocitos/fisiología , Condrocitos/ultraestructura , Colágeno Tipo II/metabolismo , Factor I del Crecimiento Similar a la Insulina/deficiencia , Ratones , Ratones Noqueados , Microscopía Electrónica , Proteína Relacionada con la Hormona Paratiroidea/biosíntesis , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Endocrinol ; 189(2): 279-87, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648295

RESUMEN

Parathyroid hormone (PTH) exerts both catabolic and anabolic actions on bone. Studies on the skeletal effects of PTH have seldom considered the effects of gender. Our study was designed to determine whether the response of mouse bone to PTH differed according to sex. As a first step, we analyzed gender differences with respect to bone mass and structural properties of 4 month old PTH treated (80 microg/kg per day for 2 weeks) male and female CD-1 mice. PTH significantly increased fat free weight/body weight, periosteal bone formation rate, mineral apposition rate, and endosteal single labeling surface, while significantly decreasing medullary area in male mice compared with vehicle treated controls, but induced no significant changes in female mice. We then analyzed the gender differences in bone marrow stromal cells (BMSC) isolated from 4 month old male and female CD-1 mice following treatment with PTH (80 microg/kg per day for 2 weeks). PTH significantly increased the osteogenic colony number and the alkaline phosphatase (ALP) activity (ALP/cell) by day 14 in cultures of BMSCs from male and female mice. PTH also increased the mRNA level of receptor activator of nuclear factor kappaB ligand in the bone tissue (marrow removed) of both females and males. However, PTH increased the mRNA levels of IGF-I and IGF-IR only in the bones of male mice. Our results indicate that on balance a 2-weeks course of PTH is anabolic on cortical bone in this mouse strain. These effects are more evident in the male mouse. These differences between male and female mice may reflect the greater response to PTH of IGF-I and IGF-IR gene expression in males enhancing the anabolic effect on cortical bone.


Asunto(s)
Huesos/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hormona Paratiroidea/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/análisis , Densidad Ósea/efectos de los fármacos , Densidad Ósea/fisiología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/metabolismo , Huesos/anatomía & histología , Huesos/efectos de los fármacos , Proteínas Portadoras/análisis , Recuento de Células/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Glicoproteínas de Membrana/análisis , Ratones , Ratones Endogámicos , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Hormona Paratiroidea/farmacología , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptor IGF Tipo 1/análisis , Factores Sexuales , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Células del Estroma/metabolismo , Tibia
16.
J Biomed Mater Res A ; 77(4): 688-99, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16514602

RESUMEN

This study was designed to test the hypothesis that the mechanical properties of a trabecular bone substitute can be enhanced through in vitro tissue formation. Our specific objectives were to (1) determine the effects of in vitro marrow stromal cell-mediated tissue deposition upon a trabeculated hydroxyapatite scaffold on the strength and toughness of the resulting bone substitute; and (2) identify and characterize regions of newly deposited matrix and mineral. This work provides a basis for future investigations aimed at transforming a brittle hydroxyapatite scaffold into an osteoinductive, biomechanically functional implant through in vitro bone deposition. As hypothesized, the mechanical properties of the trabecular bone substitutes were significantly enhanced by in vitro tissue formation. As a result of cell seeding and a 5 week culture protocol, mean strength increased by 85% (p = 0.008) and energy to fracture increased by 130% (p = 0.003). Accompanying the enhancement of mechanical properties was the deposition of significant amounts of bone matrix and mineral. Fluorescence imaging, scanning electron microscopy, electron probe microanalysis, and nanoindentation confirmed the presence of bonelike mineral with Ca/P ratio, modulus, and hardness similar to that within human and rat trabecular bone tissue. This new mineralization was found to exist within a newly deposited parallel-fibered matrix both encasing and bridging between scaffold trabeculae. Taken as a whole, our results establish the feasibility of the production of an osteoinductive hydroxyapatite-based trabecular bone substitute with mechanical properties enhanced through in vitro bone deposition.


Asunto(s)
Regeneración Ósea/fisiología , Sustitutos de Huesos , Durapatita , Ensayo de Materiales , Animales , Células de la Médula Ósea , Células Cultivadas , Humanos , Ratas , Ratas Sprague-Dawley , Células del Estroma
17.
J Nutr Biochem ; 34: 73-82, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27239754

RESUMEN

Nutrition is an important determinant of bone health and attainment of peak bone mass. Diets containing dried plum (DP) have been shown to increase bone volume and strength. These effects may be linked to the immune system and DP-specific polyphenols. To better understand these relationships, we studied DP in skeletally mature (6-month-old) and growing (1- and 2-month-old) C57Bl/6 male mice. In adult mice, DP rapidly (<2 weeks) increased bone volume (+32%) and trabecular thickness (+24%). These changes were associated with decreased osteoclast surface (Oc.S/BS) and decreased serum CTX, a marker of bone resorption. The reduction in Oc.S/BS was associated with a reduction in the osteoclast precursor pool. Osteoblast surface (Ob.S/BS) and bone formation rate were also decreased suggesting that the gain in bone in adult mice is a consequence of diminished bone resorption and formation, but resorption is reduced more than formation. The effects of DP on bone were accompanied by a decline in interleukins, TNF and MCP-1, suggesting that DP is acting in part through the immune system to suppress inflammatory activity and reduce the size of the osteoclast precursor pool. Feeding DP was accompanied by an increase in plasma phenolics, some of which have been shown to stimulate bone accrual. In growing and young adult mice DP at levels as low as 5% of diet (w/w) increased bone volume. At higher levels (DP 25%), bone volume was increased by as much as 94%. These data demonstrate that DP feeding dramatically increases peak bone mass during growth.


Asunto(s)
Desarrollo Óseo , Resorción Ósea/prevención & control , Citocinas/antagonistas & inhibidores , Alimentos en Conserva , Frutas , Alimentos Funcionales , Prunus domestica , Animales , Biomarcadores/sangre , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Remodelación Ósea , Resorción Ósea/inmunología , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/citología , Huesos/inmunología , Huesos/metabolismo , Huesos/patología , Células Cultivadas , Colágeno Tipo I/sangre , Citocinas/sangre , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/inmunología , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/citología , Osteoclastos/inmunología , Osteoclastos/metabolismo , Osteoclastos/patología , Fragmentos de Péptidos/sangre , Péptidos/sangre
18.
J Bone Miner Res ; 20(9): 1659-68, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16059637

RESUMEN

UNLABELLED: Stromal/osteoblastic cell expression of RANKL and M-CSF regulates osteoclastogenesis. We show that aging is accompanied by increased RANKL and M-CSF expression, increased stromal/osteoblastic cell-induced osteoclastogenesis, and expansion of the osteoclast precursor pool. These changes correlate with age-related alterations in the relationship between osteoblasts and osteoclasts in cancellous bone. INTRODUCTION: Bone mass is maintained through a balance between osteoblast and osteoclast activity. Osteoblasts regulate the number and activity of osteoclasts through expression of RANKL, osteoprotegerin (OPG), and macrophage-colony stimulation factor (M-CSF). To determine whether age-related changes in stromal/osteoblastic cell expression of RANKL, OPG, and M-CSF are associated with stimulation of osteoclastogenesis and whether the osteoclast precursor pool changes with age, we studied cultures of stromal/osteoblastic cells and osteoclast precursor cells from animals of different ages and examined how aging influences bone cell populations in vivo. MATERIALS AND METHODS: Osteoclast precursors from male C57BL/6 mice of 6 weeks (young), 6 months (adult), and 24 months (old) of age were either co-cultured with stromal/osteoblastic cells from young, adult, or old mice or treated with M-CSF, RANKL, and/or OPG. Osteoclast precursor pool size was determined by fluorescence-activated cell sorting (FACS), and osteoclast formation was assessed by measuring the number of multinucleated TRACP(+) cells and pit formation. The levels of mRNA for RANKL, M-CSF, and OPG were determined by quantitative RT-PCR, and transcription was measured by PCR-based run-on assays. Osteoblast and osteoclast numbers in bone were measured by histomorphometry. RESULTS: Osteoclast formation increased dramatically when stromal/osteoblastic cells from old compared with young donors were used to induce osteoclastogenesis. Regardless of the origin of the stromal/osteoblastic cells, the number of osteoclasts formed from the nonadherent population of cells increased with increasing age. Stromal/osteoblastic cell expression of RANKL and M-CSF increased, whereas OPG decreased with aging. Exogenously administered RANKL and M-CSF increased, dose-dependently, osteoclast formation from all donors, but the response was greater in cells from old donors. Osteoclast formation in vitro positively, and the ratio of osteoblasts to osteoclasts in vivo negatively, correlated with the ratio of RANKL to OPG expression in stromal/osteoblastic cells for all ages. The effects of RANKL-induced osteoclastogenesis in vitro were blocked by OPG, suggesting a causal relationship between RANKL expression and osteoclast-inducing potential. The osteoclast precursor pool and expression of RANK and c-fms increased with age. CONCLUSIONS: Our results show that aging significantly increases stromal/osteoblastic cell-induced osteoclastogenesis, promotes expansion of the osteoclast precursor pool and alters the relationship between osteoblasts and osteoclasts in cancellous bone.


Asunto(s)
Envejecimiento , Regulación de la Expresión Génica , Osteoblastos/citología , Células del Estroma/citología , Fosfatasa Ácida/farmacología , Factores de Edad , Animales , Resorción Ósea , Huesos/metabolismo , Proteínas Portadoras/biosíntesis , Núcleo Celular/metabolismo , Separación Celular , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Glicoproteínas/metabolismo , Separación Inmunomagnética , Isoenzimas/farmacología , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/citología , Masculino , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Osteoporosis , Osteoprotegerina , Reacción en Cadena de la Polimerasa , Ligando RANK , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fosfatasa Ácida Tartratorresistente , Factores de Tiempo
19.
J Bone Miner Res ; 20(1): 30-40, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15619667

RESUMEN

UNLABELLED: HA activates CD44 to stimulate RANKL expression in bone marrow stromal cells. HA stimulation of RANKL is blocked by anti-CD44 antibody and is absent in cells from CD44(-/-) mice. CD44(-/-) mice exhibit thicker cortical bone and a smaller medullary cavity, but indices of bone resorption are not affected. INTRODUCTION: Hyaluronan (HA), the major nonprotein glycosaminoglycan component of the extracellular matrix in mammalian bone marrow, functions in part through its receptor, CD44, to stimulate a series of intracellular signaling events that lead to cell migration, adhesion, and activation. To determine whether HA activation of CD44 influences RANKL and osteoprotegerin (OPG) expression and whether CD44 is functionally important in bone metabolism, we studied whole bone and bone marrow stromal cells (BMSCs) from wildtype and CD44(-/-) mice. MATERIALS AND METHODS: BMSCs from wildtype and CD44(-/-) mice at 7 weeks of age were cultured and treated with either HA or anti-CD44 antibody. The levels of mRNA of RANKL, OPG, CD44, alkaline phosphatase (ALP), osteocalcin (OC), and alphaI collagen (COLL) were determined by quantitative real-time RT-PCR. Levels of RANKL and CD44 protein were measured by immunoblotting, and expression of CD44 in whole bone was determined by immunohistochemical staining. Double immunofluorescence staining and confocal microscopy were used to study colocalization of Cbfa1, CD44, and HA. Tibias were imaged using muCT, and cancellous and cortical parameters were measured. Osteoblast and osteoclast surface in the distal femoral metaphysis and osteoclast on the endocortical surface at the tibio-fibular junction were measured using quantitative histomorphometry. Differences were analyzed using ANOVA and the Newman-Keuls test. RESULTS: Addition of HA dose-dependently increased RANKL mRNA (3.6-fold) and protein (3-fold) levels in BMSCs. Stimulation of RANKL by HA could be blocked with anti-CD44 antibody. Treatment of cells with HA or anti-CD44 antibody had no significant effect on OPG mRNA levels. Both CD44 and HA localized on the plasma membrane in cells expressing Cbfa1. HA localization on the cell membrane disappeared when cells were preincubated with anti-CD44 antibody. Compared with control mice, cortical bone of CD44(-/-) was thicker, and medullary area was smaller at both 7 and 17 weeks, but at 7 weeks, indices of bone resorption were normal. At 17 weeks of age, tibial mass of CD44(-/-) mice was higher than control mice. CD44(-/-) animals expressed less RANKL in whole bone (-30%) and in BMSCs (-50%). Cells from CD44(-/-) animals failed to respond to either HA or CD44 antibody treatment. CONCLUSIONS: HA can increase RANKL expression in BMSCs through CD44.


Asunto(s)
Células de la Médula Ósea/citología , Proteínas Portadoras/metabolismo , Receptores de Hialuranos/fisiología , Ácido Hialurónico/fisiología , Glicoproteínas de Membrana/metabolismo , Regulación hacia Arriba , Animales , Anticuerpos/farmacología , Células de la Médula Ósea/química , Células de la Médula Ósea/metabolismo , Huesos/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Receptores de Hialuranos/análisis , Receptores de Hialuranos/genética , Ácido Hialurónico/análisis , Ácido Hialurónico/farmacología , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Mutación/genética , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/metabolismo , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral , Células del Estroma/química , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo
20.
J Orthop Res ; 33(8): 1212-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25773760

RESUMEN

Available evidence indicates that some Tie2-expressing (Tie2(+) ) cells serve as multipotent progenitors that have robust BMP-dependent osteogenic activity and mediate heterotopic ossification (HO). Since signaling through the G protein Gi is required for cell motility, we hypothesized that blockade of endogenous Gi signaling in Tie2(+) cell populations would prevent HO formation. Blockade of Gi signaling in Tie2(+) cells was accomplished in transgenic mice with expression of pertussis toxin (PTX) under the control of the Tie2 promoter (Tie2(+) /PTX(+) ). Bone formation within HOs was evaluated 2 weeks after BMP injection. Expression of PTX in Tie2(+) cells significantly reduced the bone volume (BV) of HOs in male and female mice. Orthotopic bones were assessed at the distal femur and expression of PTX significantly increased trabecular bone fractional volume and bone formation rate in females only. In adult Tie2(+) /GFP(+) mice, GFP(+) cells appeared both inside and at the surfaces of bone tissue within HOs and in orthotopic bones. In summary, blockade of Gi signaling in Tie2(+) cells reduced the accrual of HOs and stimulated osteogenesis in orthotopic bones. Targeting of Gi protein coupled receptors in Tie2(+) cells may be a novel therapeutic strategy in states of abnormal bone formation such as osteoporosis and HO.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Osificación Heterotópica/fisiopatología , Osteogénesis , Receptor TIE-2/análisis , Transducción de Señal/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos , Caracteres Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA