Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Osteoarthritis Cartilage ; 27(10): 1557-1563, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31176017

RESUMEN

OBJECTIVE: To investigate the impact of deleting Suppressor of Cytokine Signaling (SOCS)-3 (SOCS3) in chondrocytes during murine skeletal development. METHOD: Mice with a conditional Socs3 allele (Socs3fl/fl) were crossed with a transgenic mouse expressing Cre recombinase under the control of the type II collagen promoter (Col2a1) to generate Socs3Δ/Δcol2 mice. Skeletal growth was analyzed over the lifespan of Socs3Δ/Δcol2 mice and controls by detailed histomorphology. Bone size and cortical bone development was evaluated by micro-computed tomography (micro-CT). Growth plate (GP) zone width, chondrocyte proliferation and apoptosis were assessed by immunofluorescence staining for Ki67 and TUNEL. Fibroblast growth factor receptor-3 (FGFR3) signaling in the GP was assessed by immunohistochemistry, while the effect of SOCS3 overexpression on FGFR3-driven pMAPK signaling in HEK293T cells was evaluated by Western blot. RESULTS: Socs3Δ/Δcol2 mice of both sexes were consistently smaller compared to littermate controls throughout life. This phenotype was due to reduced long bone size, poor cortical bone development, reduced Ki67+ proliferative chondrocytes and decreased proliferative zone (PZ) width in the GP. Expression of pMAPK, but not pSTAT3, was increased in the GPs of Socs3Δ/Δcol2 mice relative to littermate controls. Overexpression of FGFR3 in HEK293T cells increased Fibroblast Growth Factor 18 (FGF18)-dependent Mitogen-activated protein kinase (MAPK) phosphorylation, while concomitant expression of SOCS3 reduced FGFR3 expression and abrogated MAPK signaling. CONCLUSION: Our results suggest a potential role for SOCS3 in GP chondrocyte proliferation by regulating FGFR3-dependent MAPK signaling in response to FGF18.


Asunto(s)
Desarrollo Óseo/fisiología , Condrocitos/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteína 1 Supresora de la Señalización de Citocinas/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal
2.
Scand J Rheumatol ; 44(3): 182-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25439190

RESUMEN

OBJECTIVES: Progressive destruction of synovial joint cartilage and bone occurs in pathological conditions such as rheumatoid arthritis (RA) because of the overproduction of pro-inflammatory cytokines and activation of nuclear factor kappa B (NF-κB). Through the screening of NF-κB inhibitors by a luciferase reporter gene assay, we identified parthenolide (PAR) as the most potent NF-κB inhibitor, among several PAR analogue compounds. This study was undertaken to determine whether PAR inhibits pro-inflammatory cytokine production, cartilage degradation, and inflammatory arthritis. METHOD: The mRNA levels of pro-inflammatory cytokines were examined by real-time polymerase chain reaction (PCR). Proteoglycan content and release were determined by measuring glycosaminoglycan (GAG) levels using the dimethylmethylene blue (DMMB) dye-binding assay. The potential role of PAR in treatment of arthritis was studied using a collagen-induced arthritis (CIA) model. RESULTS: We established that PAR, as a prototype compound, suppressed lipopolysaccharide (LPS)- and tumour necrosis factor (TNF)-α-induced increases in matrix metalloproteinase (MMP)-1, MMP-3, inducible nitric oxide synthase (iNOS), and interleukin (IL)-1ß mRNA in chondrocytes. In addition, PAR prevented proteoglycan degradation triggered by pro-inflammatory cytokines. PAR treatment at the onset of CIA symptoms significantly reduced synovitis, inflammation, and pannus formation scores. Reduced synovial inflammation after PAR treatment was also reflected in significantly less bone erosion and cartilage damage. CONCLUSIONS: These data indicate a protective effect of PAR on the catabolic insults of pro-inflammatory cytokines on chondrocyte metabolism and GAG release in vitro and in CIA. PAR had anti-inflammatory and structure-modifying effects on experimental arthritis, suggesting that PAR may be useful as a potential alternative or adjunct therapy for inflammatory arthritis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Artritis Experimental , Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Citocinas/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , ARN Mensajero/efectos de los fármacos , Sesquiterpenos/farmacología , Membrana Sinovial/efectos de los fármacos , Animales , Cartílago Articular/patología , Condrocitos/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Metaloproteinasa 1 de la Matriz/efectos de los fármacos , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/efectos de los fármacos , Metaloproteinasa 3 de la Matriz/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Mensajero/metabolismo , Ratas , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/farmacología
3.
Dev Biol ; 344(2): 1001-10, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20599900

RESUMEN

Bone development is dependent on the functionality of three essential cell types: chondrocytes, osteoclasts and osteoblasts. If any of these cell types is dysfunctional, a developmental bone phenotype can result. The bone disease osteopetrosis is caused by osteoclast dysfunction or impaired osteoclastogenesis, leading to increased bone mass. In ClC-7 deficient mice, which display severe osteopetrosis, the osteoclast malfunction is due to abrogated acidification of the resorption lacuna. This study sought to investigate the consequences of osteoclast malfunction on bone development, bone structure and bone modeling/remodeling in ClC-7 deficient mice. Bones from wildtype, heterozygous and ClC-7 deficient mice were examined by bone histomorphometry and immunohistochemistry. ClC-7 deficient mice were found to have a severe developmental bone phenotype, characterized by dramatically increased bone mass, a high content of cartilage remnants, impaired longitudinal and radial growth, as well as lack of compact cortical bone development. Indices of bone formation were reduced in ClC-7 deficient mice; however, calcein labeling indicated that mineralization occurred on most trabecular bone surfaces. Osteoid deposition had great regional variance, but an osteopetrorickets phenotype, as observed in oc/oc mice, was not apparent in the ClC-7 deficient mice. A striking finding was the presence of very large abnormal osteoclasts, which filled the bone marrow space within the ClC-7 deficient bones. The development of these giant osteoclasts could be due to altered cell fate of the ClC-7 deficient osteoclasts, caused by increased cellular fusion and/or prolonged osteoclast survival. In summary, malfunctional ClC-7 deficient osteoclasts led to a severe developmental bone phenotype including abnormally large and non-functional osteoclasts. Bone formation paremeters were reduced; however, bone formation and mineralization were found to be heterogenous and continuing.


Asunto(s)
Huesos/fisiopatología , Animales , Desarrollo Óseo/genética , Matriz Ósea/fisiopatología , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/fisiopatología , Cartílago/fisiopatología , Comunicación Celular , Diferenciación Celular , Citocinas , Homocigoto , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis , Osteopetrosis/genética , Osteopetrosis/metabolismo
4.
Nat Med ; 6(9): 985-90, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10973317

RESUMEN

Members of the AP-1 family of transcription factors participate in the regulation of bone cell proliferation and differentiation. We report here a potent AP-1-related regulator of osteoblast function: DeltaFosB, a naturally occurring truncated form of FosB that arises from alternative splicing of the fosB transcript and is expressed in osteoblasts. Overexpression of DeltaFosB in transgenic mice leads to increased bone formation throughout the skeleton and a continuous post-developmental increase in bone mass, leading to osteosclerosis. In contrast, DeltaFosB inhibits adipogenesis both in vivo and in vitro, and downregulates the expression of early markers of adipocyte differentiation. Because osteoblasts and adipocytes are thought to share a common precursor, it is concluded that DeltaFosB transcriptionally regulates osteoblastogenesis, possibly at the expense of adipogenesis.


Asunto(s)
Adipocitos/citología , Calcinosis/genética , Osteoblastos/citología , Osteosclerosis/genética , Proteínas Proto-Oncogénicas c-fos/genética , Empalme Alternativo , Animales , Antígenos de Diferenciación , Densidad Ósea , Diferenciación Celular , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/biosíntesis , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas c-fos/biosíntesis
5.
Adv Exp Med Biol ; 658: 51-60, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19950015

RESUMEN

Members of the ephrin and Eph family are local mediators of cell function through largely contact-dependent processes in development and in maturity. Production of ephrinB2 mRNA and protein are increased by PTH and PTHrP in osteoblasts. Both a synthetic peptide antagonist of ephrinB2/EphB4 receptor interaction and recombinant soluble extracellular domain of EphB4 (sEphB4), which is an antagonist of both forward and reverse EphB4 signaling, were able to inhibit mineralization and the expression of several osteoblast genes involved late in osteoblast differentiation. The findings are consistent with ephrinB2/EphB4 signaling within the osteoblast lineage having a paracrine role in osteoblast differentiation, in addition to the proposed role of osteoclast-derived ephrinB2 in coupling of bone formation to resorption. This local regulation might contribute to control of osteoblast differentiation and bone formation at remodeling sites, and perhaps also in modeling.


Asunto(s)
Linaje de la Célula , Efrina-B2/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Receptor EphB4/metabolismo , Transducción de Señal , Animales , Comunicación Celular , Humanos , Ratones , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis , Ratas
6.
J Cell Biol ; 151(2): 311-20, 2000 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-11038178

RESUMEN

c-src deletion in mice leads to osteopetrosis as a result of reduced bone resorption due to an alteration of the osteoclast. We report that deletion/reduction of Src expression enhances osteoblast differentiation and bone formation, contributing to the increase in bone mass. Bone histomorphometry showed that bone formation was increased in Src null compared with wild-type mice. In vitro, alkaline phosphatase (ALP) activity and nodule mineralization were increased in primary calvarial cells and in SV40-immortalized osteoblasts from Src(-/-) relative to Src(+/+) mice. Src-antisense oligodeoxynucleotides (AS-src) reduced Src levels by approximately 60% and caused a similar increase in ALP activity and nodule mineralization in primary osteoblasts in vitro. Reduction in cell proliferation was observed in primary and immortalized Src(-/-) osteoblasts and in normal osteoblasts incubated with the AS-src. Semiquantitative reverse transcriptase-PCR revealed upregulation of ALP, Osf2/Cbfa1 transcription factor, PTH/PTHrP receptor, osteocalcin, and pro-alpha 2(I) collagen in Src-deficient osteoblasts. The expression of the bone matrix protein osteopontin remained unchanged. Based on these results, we conclude that the reduction of Src expression not only inhibits bone resorption, but also stimulates osteoblast differentiation and bone formation, suggesting that the osteogenic cells may contribute to the development of the osteopetrotic phenotype in Src-deficient mice.


Asunto(s)
Proteínas de Neoplasias , Osteoblastos/citología , Osteogénesis/genética , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Fosfatasa Alcalina/biosíntesis , Animales , Resorción Ósea/genética , Diferenciación Celular , División Celular , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Mutantes , Oligonucleótidos Antisentido/farmacología , Osteopetrosis/genética , Hormona Paratiroidea/biosíntesis , Fenotipo , Receptores de Hormona Paratiroidea/biosíntesis , Cráneo/citología , Factores de Transcripción/biosíntesis , Transcripción Genética
7.
Matrix Biol ; 83: 77-96, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31381970

RESUMEN

Cartilage remodelling and chondrocyte differentiation are tightly linked to angiogenesis during bone development and endochondral ossification. To investigate whether collagenase-mediated cleavage of the major cartilage collagen (collagen II) plays a role in this process, we generated a knockin mouse in which the mandatory collagenase cleavage site at PQG775↓776LAG, was mutated to PPG775↓776MPG (Col2a1Bailey). This approach blocked collagen II cleavage, and the production of putative collagen II matrikines derived from this site, without modifying matrix metalloproteinase expression or activity. We report here that this mouse (Bailey) is viable. It has a significantly expanded growth plate and exhibits delayed and abnormal angiogenic invasion into the growth plate. Deeper electron microscopy analyses revealed that, at around five weeks of age, a small number of blood vessel(s) penetrate into the growth plate, leading to its abrupt shrinking and the formation of a bony bridge. Our results from in vitro and ex vivo studies suggest that collagen II matrikines stimulate the normal branching of endothelial cells and promote blood vessel invasion at the chondro-osseous junction. The results further suggest that failed collagenolysis in Bailey leads to expansion of the hypertrophic zone and formation of a unique post-hypertrophic zone populated with chondrocytes that re-enter the cell cycle and proliferate. The biological rescue of this in vivo phenotype features the loss of a substantial portion of the growth plate through aberrant ossification, and narrowing of the remaining portion that leads to limb deformation. Together, these data suggest that collagen II matrikines stimulate angiogenesis in skeletal growth and development, revealing novel strategies for stimulating angiogenesis in other contexts such as fracture healing and surgical applications.


Asunto(s)
Condrocitos/citología , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Colagenasas/metabolismo , Placa de Crecimiento/anomalías , Animales , Diferenciación Celular , Proliferación Celular , Colágeno Tipo II/química , Femenino , Técnicas de Sustitución del Gen , Placa de Crecimiento/irrigación sanguínea , Masculino , Ratones , Neovascularización Fisiológica , Osteogénesis
8.
Equine Vet J ; 50(2): 255-260, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28833497

RESUMEN

BACKGROUND: Palmar/plantar osteochondral disease (POD) and third metacarpal/-tarsal condylar fractures are considered fatigue injuries of subchondral bone (SCB) and calcified cartilage due to repetitive high loads in racehorses. In combination with adaptive changes in SCB in response to race training, the accumulation of SCB fatigue is likely to result in changes of joint surface mechanical properties. OBJECTIVES: To determine the spatial relationship and correlation of calcified articular surface biomechanical properties with SCB microstructure and training history in the distal palmar metacarpal condyle of Thoroughbred racehorses. STUDY DESIGN: Cross-sectional study. METHODS: Third metacarpal condyles were examined from 31 Thoroughbred horses with micro-computed tomography (microCT). Hyaline cartilage was removed and reference point indentation (RPI) mechanical testing of the calcified articular surface was performed. Training histories were obtained from trainers. The association among indentation distance increase (IDI, an inverse RPI measure of toughness), and microCT and training variables was assessed using a mixed-effects generalised linear model. RESULTS: Untrained horses had higher IDI than horses that had commenced training (P<0.001). Death as a result of musculoskeletal bone fatigue injury (P = 0.044) and presence of POD (P = 0.05) were associated with higher IDI. The microCT variables connectivity density and trabecular pattern factor were positively (P = 0.002) and negatively (P<0.001) correlated with IDI respectively. MAIN LIMITATIONS: The application of RPI to the calcified articular surface is novel and there is a potential for measurement variability with surface unevenness. CONCLUSION: Commencement of race training is associated with altered material properties of the calcified articular surface in horses. Reduced articular surface material properties can also be detected in horses that have fatigue injuries of the distal metacarpus and at other sites in the skeleton. Measures of SCB connectivity and trabecular surface shape may be more important determinants of resistance to failure of the calcified articular surface than traditional measures such as SCB volume and density.


Asunto(s)
Densidad Ósea/fisiología , Huesos/anatomía & histología , Caballos , Huesos del Metacarpo/fisiología , Animales , Fenómenos Biomecánicos , Cartílago Articular , Estudios Transversales , Condicionamiento Físico Animal , Deportes , Microtomografía por Rayos X
9.
J Clin Invest ; 106(9): 1095-103, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11067862

RESUMEN

Growth hormone (GH) regulates both bone growth and remodeling, but it is unclear whether these actions are mediated directly by the GH receptor (GHR) and/or IGF-I signaling. The actions of GH are transduced by the Jak/Stat signaling pathway via Stat5, which is thought to regulate IGF-I expression. To determine the respective roles of GHR and IGF-I in bone growth and remodeling, we examined bones of wild-type, GHR knockout (GHR(-/-)), Stat5ab(-/-), and GHR(-/-) mice treated with IGF-I. Reduced bone growth in GHR(-/-) mice, due to a premature reduction in chondrocyte proliferation and cortical bone growth, was detected after 2 weeks of age. Additionally, although trabecular bone volume was unchanged, bone turnover was significantly reduced in GHR(-/-) mice, indicating GH involvement in the high bone-turnover level during growth. IGF-I treatment almost completely rescued all effects of the GHR(-/-) on both bone growth and remodeling, supporting a direct effect of IGF-I on both osteoblasts and chondrocytes. Whereas bone length was reduced in Stat5ab(-/-) mice, there was no reduction in trabecular bone remodeling or growth-plate width as observed in GHR(-/-) mice, indicating that the effects of GH in bone may not involve Stat5 activation.


Asunto(s)
Desarrollo Óseo/fisiología , Remodelación Ósea/fisiología , Hormona del Crecimiento/deficiencia , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas de la Leche , Animales , Desarrollo Óseo/efectos de los fármacos , Desarrollo Óseo/genética , Remodelación Ósea/efectos de los fármacos , Remodelación Ósea/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Hormona del Crecimiento/genética , Hormona del Crecimiento/fisiología , Homeostasis , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT5 , Transactivadores/deficiencia , Transactivadores/genética , Transactivadores/fisiología
10.
J Clin Invest ; 107(3): 277-86, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11160151

RESUMEN

Parathyroid hormone (PTH), an important regulator of calcium homeostasis, targets most of its complex actions in bone to cells of the osteoblast lineage. Furthermore, PTH is known to stimulate osteoclastogenesis indirectly through activation of osteoblastic cells. To assess the role of the PTH/PTH-related protein receptor (PPR) in mediating the diverse actions of PTH on bone in vivo, we generated mice that express, in cells of the osteoblastic lineage, one of the constitutively active receptors described in Jansen's metaphyseal chondrodysplasia. In these transgenic mice, osteoblastic function was increased in the trabecular and endosteal compartments, whereas it was decreased in the periosteum. In trabecular bone of the transgenic mice, there was an increase in osteoblast precursors, as well as in mature osteoblasts. Osteoblastic expression of the constitutively active PPR induced a dramatic increase in osteoclast number in both trabecular and compact bone in transgenic animals. The net effect of these actions was a substantial increase in trabecular bone volume and a decrease in cortical bone thickness of the long bones. These findings, for the first time to our knowledge, identify the PPR as a crucial mediator of both bone-forming and bone-resorbing actions of PTH, and they underline the complexity and heterogeneity of the osteoblast population and/or their regulatory microenvironment.


Asunto(s)
Remodelación Ósea , Huesos/metabolismo , Osteoblastos/metabolismo , Hormona Paratiroidea/fisiología , Receptores de Hormona Paratiroidea/genética , Factores de Edad , Animales , Huesos/citología , Huesos/efectos de los fármacos , Hibridación in Situ , Ratones , Ratones Transgénicos , Mutación , Osteoblastos/efectos de los fármacos , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/biosíntesis , Transducción de Señal , Tibia/citología , Tibia/efectos de los fármacos , Tibia/metabolismo
11.
Ann N Y Acad Sci ; 1068: 458-70, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16831943

RESUMEN

Since parathyroid hormone (PTH) is the only proven anabolic therapy for bone, it becomes the benchmark by which new treatments will be evaluated. The anabolic effect of PTH is dependent upon intermittent administration, but when an elevated PTH level is maintained even for a few hours it initiates processes leading to new osteoclast formation, and the consequent resorption overrides the effects of activating genes that direct bone formation. Identification of PTH-related protein (PTHrP) production by cells early in the osteoblast lineage, and its action through the PTH1R upon more mature osteoblastic cells, together with the observation that PTHrP+/- mice are osteoporotic, all raise the possibility that PTHrP is a crucial paracrine regulator of bone formation. The finding that concurrent treatment with bisphosphonates impairs the anabolic response to PTH, adds to other clues that osteoclast activity is necessary to complement the direct effect that PTH has in promoting differentiation of committed osteoblast precursors. This might involve the generation of a coupling factor from osteoclasts that are transiently activated by receptor activator of nuclear factor-kappaB ligand (RANKL) in response to PTH. New approaches to anabolic therapies may come from the discovery that an activating mutation in the LRP5 gene is responsible for an inherited high bone mass syndrome, and the fact that this can be recapitulated in transgenic mice, whereas inactivating mutations result in severe bone loss. This has focused attention on the Wnt/frizzled/beta-catenin pathway as being important in bone formation, and proof of the concept has been obtained in experimental models.


Asunto(s)
Anabolizantes/uso terapéutico , Desarrollo Óseo/fisiología , Enfermedades Óseas/tratamiento farmacológico , Animales , Humanos , Ratones , Ratones Noqueados , Hormona Paratiroidea/fisiología , Receptores de Hormona Paratiroidea/fisiología , Transducción de Señal
12.
Mol Endocrinol ; 11(11): 1695-708, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9328351

RESUMEN

Human and murine osteocalcin genes demonstrate similar cell-specific expression patterns despite significant differences in gene locus organization and sequence variations in cis-acting regulatory elements. To investigate whether differences in these regulatory regions result in an altered response to 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3] in vivo, we compared the response of the endogenous mouse osteocalcin gene to a bacterial reporter gene directed by flanking regions of the human osteocalcin gene in transgenic mice. Transgene expression colocalized with endogenous osteocalcin expression in serial sections, being detected in osteoblasts, osteocytes and hypertrophic chondrocytes. In calvarial cell culture lysates from transgenic and nontransgenic mice, the endogenous mouse osteocalcin gene did not respond to 1,25-(OH)2D3 treatment. Despite this, transgene activity was significantly increased in the same cells. Similarly, Northern blots of total cellular RNA and in situ hybridization studies of transgenic animals demonstrated a maximal increase in transgene expression at 6 h after 1,25-(OH)2D3 injection (23.6+/-3.6-fold) with a return to levels equivalent to uninjected animals by 24 h (1.2+/-0.1-fold). This increase in transgene expression was also observed at 6 h after 1,25-(OH)2D3 treatment in animals on a low calcium diet (25.2+/-7.7-fold) as well as in transgenic mice fed a vitamin D-deficient diet containing strontium chloride to block endogenous 1,25-(OH)2D3 production (7.5+/-0.9-fold). In contrast to the increased transgene expression levels, neither endogenous mouse osteocalcin mRNA levels nor serum osteocalcin levels were significantly altered after 1,25-(OH)2D3 injection in transgenic or nontransgenic mice, regardless of dietary manipulations, supporting evidence for different mechanisms regulating the response of human and mouse osteocalcin genes to 1,25-(OH)2D3. Although the cis- and trans-acting mechanisms directing cell-specific gene expression appear to be conserved in the mouse and human osteocalcin genes, responsiveness to 1,25-(OH)2D3 is not. The mouse osteocalcin genes do not respond to 1,25-(OH)2D3 treatment, but the human osteocalcin-directed transgene is markedly upregulated under the same conditions and in the same cells. The divergent responses of these homologous genes to 1,25-(OH)2D3 are therefore likely to be due to differences in mouse and human osteocalcin-regulatory sequences rather than to variation in the complement of trans-acting factors present in mouse osteoblastic cells. Increased understanding of these murine-human differences in osteocalcin regulation may shed light on the function of osteocalcin and its regulation by vitamin D in bone physiology.


Asunto(s)
Huesos/metabolismo , Calcitriol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Osteocalcina/biosíntesis , Animales , Huesos/citología , Calcio/deficiencia , Calcio de la Dieta/farmacología , Cartílago/citología , Cartílago/metabolismo , Fémur/citología , Fémur/metabolismo , Genes Reporteros , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos , Ratones Transgénicos , Especificidad de Órganos , Osteoblastos/metabolismo , Osteocalcina/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Cráneo/citología , Cráneo/metabolismo , Especificidad de la Especie , Estroncio/toxicidad , Transgenes/efectos de los fármacos , Deficiencia de Vitamina D/genética , Deficiencia de Vitamina D/metabolismo
13.
J Bone Miner Res ; 9(12): 1859-63, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7872050

RESUMEN

The evidence for a role of parathyroid hormone in the bone loss after the menopause remains controversial. This study examines the effect of parathyroidectomy on femoral trabecular bone volume, thickness, and spacing and biochemical markers of bone turnover in the oophorectomized rat. Female Sprague-Dawley rats 3 months old were double sham operated (sham), oophorectomized (OPX), parathyroidectomized (PTX), or oophorectomized and parathyroidectomized (O/P) under halothane anesthesia. At 9 weeks postoperation, femoral trabecular bone volume (BV/TV) was lower in OPX and O/P rats compared with sham or PTX animals (BV/TV, %, mean +/- SEM): sham 25.9 +/- 0.5, OPX 15.1 +/- 0.9, PTX 24.1 +/- 0.9, O/P 17.3 +/- 0.5; p < 0.001). Urinary hydroxyproline excretion, serum osteocalcin, and alkaline phosphatase activity were higher in OPX and O/P rats compared with control animals at 3 weeks postoperation (OHPE microM GF, mean +/- SEM: sham 1.37 +/- 0.16, OPX 2.16 +/- 0.26, PTX 0.95 +/- 0.21, O/P 1.92 +/- 0.22, p < 0.005; osteocalcin, microgram/liter, sham 31.8 +/- 1.8, OPX 33.7 +/- 2.7, PTX 24.5 +/- 2.1, O/P 34.3 +/- 2.1, p < 0.025; alkaline phosphatase, U/liter, sham 90 +/- 3, OPX 125 +/- 9, PTX 87 +/- 9, O/P 116 +/- 11, p < 0.005). These data indicate postoophorectomy bone loss is not prevented by parathyroidectomy.


Asunto(s)
Osteoporosis/prevención & control , Paratiroidectomía , Fosfatasa Alcalina/sangre , Animales , Densidad Ósea , Calcio/sangre , Modelos Animales de Enfermedad , Femenino , Fémur/fisiopatología , Hidroxiprolina/orina , Osteocalcina/sangre , Osteoporosis/sangre , Osteoporosis/fisiopatología , Osteoporosis/orina , Ovariectomía/efectos adversos , Ratas , Ratas Sprague-Dawley
14.
Endocrinology ; 143(11): 4304-9, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12399426

RESUMEN

Transgenic mice overexpressing deltaFosB, a naturally occurring splice variant of FosB, develop an osteosclerotic phenotype. The increased bone formation has been shown to be due, at least in part, to autonomous effects of deltaFosB isoforms on cells of the osteoblast lineage. However, abdominal fat and marrow adipocytes are also markedly decreased in deltaFosB mice, leading to low serum leptin levels. Increased bone mass has been linked to the absence of leptin and leptin receptor signaling in ob/ob and db/db mice. Thus, in addition to affecting directly osteoblastogenesis and bone formation, deltaFosB isoforms might increase bone mass indirectly via a decrease in leptin. To test this hypothesis, we restored normal circulating levels of leptin in deltaFosB mice via sc implanted osmotic pumps. Complete histomorphometric analysis demonstrated that trabecular bone volume as well as dynamic parameters of bone formation was unchanged by this treatment in both deltaFosB transgenic mice and control littermates. This demonstration that restoring circulating levels of leptin in deltaFosB transgenic mice failed to rescue the bone phenotype further indicates that the marked increase in bone formation is autonomous to the osteoblast lineage.


Asunto(s)
Huesos/anatomía & histología , Leptina/sangre , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/fisiología , Animales , Desarrollo Óseo , Leptina/farmacología , Ratones , Ratones Transgénicos , Osteoblastos/fisiología , Fosforilación , Fosfotirosina/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal
15.
Bone ; 19(5): 455-61, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8922643

RESUMEN

The effect of short-term estradiol treatment, administered from the time of ovariectomy, on increased bone turnover and subsequent bone loss was studied in the rat. Adult female Sprague-Dawley rats were ovariectomized and administered daily subcutaneous (s.c.) injections of 17 beta-estradiol at 8 micrograms/ kg per day (Low) and 20 micrograms/kg per day (High) or vehicle alone (Veh). Femoral trabecular bone volume (BV/TV) and trabecular number (Tb.N) in the distal femur were transiently increased at 6 days postoperation in a dose-dependent manner following estradiol administration [mean +/- SEM: BV/TV (%), day 0, 6.6 +/- 0.2; day 6, Veh 7.8 +/- 0.4, Low 10.2 +/- 2.2, High 12.8 +/- 1.7 (p < 0.05); Tb.N (/mm), day 0, 2.30 +/- 0.24; day 6, Veh 2.89 +/- 0.33, Low 3.4 +/- 0.7, High 4.39 +/- 0.34 (p < 0.05)]. Estradiol prevented the ovariectomy-induced decrease in BV/TV and Tb.N between 9 and 15 days observed in Veh rats. Both serum alkaline phosphatase and urine hydroxyproline excretion were maintained at preoperative levels or lower from day 6 postoperation with high dose estradiol. Serum osteocalcin, however, rose above preoperative levels with estradiol at days 6 and 9, but returned to these values on days 15 and 21 postoperation. These results suggest that estradiol, administered from the time of ovariectomy, immediately suppressed markers associated with osteoblast proliferation/matrix synthesis and bone resorption. Mineralization does not appear to be so rapidly suppressed by estradiol with relatively high levels immediately following administration, resulting in a transient increase in trabecular bone volume and trabecular number.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Estradiol/uso terapéutico , Osteoporosis Posmenopáusica/tratamiento farmacológico , Fosfatasa Alcalina/sangre , Análisis de Varianza , Animales , Resorción Ósea/prevención & control , División Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estradiol/administración & dosificación , Estradiol/farmacología , Terapia de Reemplazo de Estrógeno , Femenino , Fémur/efectos de los fármacos , Fémur/fisiología , Humanos , Hidroxiprolina/orina , Inyecciones Subcutáneas , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteocalcina/sangre , Osteoporosis Posmenopáusica/prevención & control , Ovariectomía/efectos adversos , Ratas , Ratas Sprague-Dawley
16.
Bone ; 30(1): 32-9, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11792562

RESUMEN

The DeltaFosB isoforms are naturally occurring AP-1 family members that increase bone volume via a cell-autonomous effect on osteoblastic bone formation. Mice overexpressing DeltaFosB demonstrate a very high level of bone formation, resulting in a progressive osteosclerosis. Despite the linkage of bone formation and resorption in physiological systems, no alteration in bone resorption was detected in mice overexpressing DeltaFosB. To determine whether altering DeltaFosB expression can regulate bone formation independently of bone resorption in adult mice, we used the Tet-Off-inducible transgene system to induce or block transgenic DeltaFosB overexpression and thereby regulate bone formation in vivo. Overexpression of DeltaFosB after skeletal maturity increased trabecular bone volume by increasing bone formation, again without altering bone resorption, indicating that developmental DeltaFosB overexpression is not required for the osteosclerotic phenotype. Similarly, switching off DeltaFosB overexpression after osteosclerosis had developed led to a marked decrease in bone formation and loss of bone mass such that trabecular bone volume approached normal levels. Despite this dramatic reduction, no alteration in bone resorption was detected. These results clearly demonstrate that DeltaFosB regulates bone formation and bone mass in adult mice with no effect on bone resorption.


Asunto(s)
Densidad Ósea/genética , Osteogénesis/genética , Proteínas Proto-Oncogénicas c-fos/genética , Animales , Resorción Ósea/genética , Doxiciclina/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Transgénicos , Osteosclerosis/genética , Osteosclerosis/prevención & control , Ovariectomía , Tetraciclina/farmacología , Factor de Transcripción AP-1/genética
17.
Bone ; 25(3): 261-7, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10495129

RESUMEN

Naturally occuring inactivating mutations of the Src homology 2 (SH2) domain-containing tyrosine phosphatase 1 (SHP-1) in mice give rise to the motheaten (me) phenotype. me/me mice have multiple hematopoietic abnormalities, suggesting that this phosphatase plays an important role in hematopoiesis. SHP-1 binds to and is activated by several hematopoietic surface receptors, including the colony-stimulating factor type 1 receptor. We have examined the role of SHP-1 in osteoclastogenesis and osteoclast function using mice with the viable motheaten (me(v)/me(v)) mutation, which has markedly decreased SHP-1 activity. Histomorphometric analysis of 6-week-old me(v)/me(v) mice and control littermates showed a marked osteopenia with an increase in bone resorption indices. The number of formed osteoclast-like cells (OCLs) in cocultures of me(v)/me(v) hematopoietic cells with normal osteoblasts was significantly increased. In contrast, the number of OCLs formed in the coculture of normal bone marrow cells with the me(v)/me(v) osteoblasts was not significantly different from controls. The bone-resorbing activity of me(v)me(v) OCLs and authentic osteoclasts was also found to be increased. Finally, Western blotting of proteins from me(v)/me(v) and control OCLs revealed an overall increase in tyrosine phosphorylation in the me(v)/me(v) lysates. These in vivo and in vitro results suggest that SHP-1 is a negative regulator of bone resorption, affecting both the formation and the function of osteoclasts.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Resorción Ósea/metabolismo , Osteoclastos/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Dominios Homologos src/fisiología , Animales , Animales Recién Nacidos , Western Blotting , Enfermedades Óseas Metabólicas/patología , Células de la Médula Ósea/enzimología , Células Cultivadas , Técnicas de Cocultivo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Osteoclastos/enzimología , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas con Dominio SH2 , Transducción de Señal , Cráneo/citología , Cráneo/enzimología , Bazo/citología , Tibia/crecimiento & desarrollo , Tibia/patología
18.
Bone ; 30(1): 18-25, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11792560

RESUMEN

To determine the contributions of estrogen receptor (ER)alpha and ERbeta in bone growth and remodeling in male and female mice, we generated and analyzed full knockouts for each receptor, and a double ER knockout. Although suppression of the ligand to the ERs (i.e., estradiol) after menopause or gonadectomy in females led to a catastrophic increase in bone turnover and concomitant bone loss, deletion of one or both ERs failed to show such an effect. Complete deletion of ERalpha led to a decrease, not an increase, in bone turnover and an increase, not a decrease, in trabecular bone volume in both male and female animals. Deletion of ERbeta led to different responses in males, where bone was unaffected, and in females, where bone resorption was decreased and trabecular bone volume increased. In contrast, deletion of both ERs led to a profound decrease in trabecular bone volume in females, which was associated with a decrease, not an increase, in bone turnover. Finally, deletion of ERalpha, but not ERbeta, led to major changes in circulating levels of estradiol and/or testosterone, indirectly affecting bone remodeling and bone mass. Thus, only ERalpha was shown to regulate bone remodeling in males, whereas in females both receptor subtypes influenced this process and could, at least under basal knockout conditions, compensate for each other.


Asunto(s)
Remodelación Ósea/fisiología , Receptores de Estrógenos/deficiencia , Receptores de Estrógenos/genética , Animales , Densidad Ósea/fisiología , Desarrollo Óseo/fisiología , Estradiol/sangre , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Masculino , Ratones , Ratones Noqueados , Caracteres Sexuales , Testosterona/sangre
20.
Bone ; 50(3): 704-12, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22173052

RESUMEN

Proteinase-activated receptor-2 (PAR(2)) is a G-protein coupled receptor expressed by osteoblasts and monocytes. PAR(2) is activated by a number of proteinases including coagulation factors and proteinases released by inflammatory cells. The aim of the current study was to investigate the role of PAR(2) in skeletal growth and repair using wild type (WT) and PAR(2) knockout (KO) mice. Micro computed tomography and histomorphometry were used to examine the structure of tibias isolated from uninjured mice at 50 and 90 days of age, and from 98-day-old mice in a bone repair model in which a hole had been drilled through the tibias. Bone marrow was cultured and investigated for the presence of osteoblast precursors (alkaline phosphatase-positive fibroblastic colonies), and osteoclasts were counted in cultures treated with M-CSF and RANKL. Polymerase chain reaction (PCR) was used to determine which proteinases that activate PAR(2) are expressed in bone marrow. Regulation of PAR(2) expression in primary calvarial osteoblasts from WT mice was investigated by quantitative PCR. Cortical and trabecular bone volumes were significantly greater in the tibias of PAR(2) KO mice than in those of WT mice at 50 days of age. In trabecular bone, osteoclast surface, osteoblast surface and osteoid volume were significantly lower in KO than in WT mice. Bone marrow cultures from KO mice showed significantly fewer alkaline phosphatase-positive colony-forming units and osteoclasts compared to cultures from WT mice. Significantly less new bone and significantly fewer osteoclasts were observed in the drill sites of PAR(2) KO mice compared to WT mice 7 days post-surgery. A number of activators of PAR(2), including matriptase and kallikrein 4, were found to be expressed by normal bone marrow. Parathyroid hormone, 1,25 dihydroxyvitamin D(3), or interleukin-6 in combination with its soluble receptor down-regulated PAR(2) mRNA expression, and fibroblast growth factor-2 or thrombin stimulated PAR(2) expression. These results suggest that PAR(2) activation contributes to determination of cells of both osteoblast and osteoclast lineages within bone marrow, and thereby participates in the regulation of skeletal growth and bone repair.


Asunto(s)
Desarrollo Óseo/fisiología , Diferenciación Celular/fisiología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptor PAR-2/metabolismo , Tibia/crecimiento & desarrollo , Animales , Calcitriol/metabolismo , Células Cultivadas , Interleucina-6/metabolismo , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoclastos/citología , Hormona Paratiroidea/metabolismo , Radiografía , Receptor PAR-2/genética , Tibia/diagnóstico por imagen , Tibia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA