Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
FASEB J ; 35(5): e21578, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33835498

RESUMEN

Bone loss is a major health concern for astronauts during long-term spaceflight and for patients during prolonged bed rest or paralysis. Growing evidence suggests that osteocytes, the most abundant cells in the mineralized bone matrix, play a key role in sensing mechanical forces applied to the skeleton and integrating the orchestrated response into subcellular biochemical signals to modulate bone homeostasis. However, the precise molecular mechanisms underlying both mechanosensation and mechanotransduction in late-osteoblast-to-osteocyte cells under microgravity (µG) have yet to be elucidated. To unravel the mechanisms by which late osteoblasts and osteocytes sense and respond to mechanical unloading, we exposed the osteocytic cell line, Ocy454, to 2, 4, or 6 days of µG on the SpaceX Dragon-6 resupply mission to the International Space Station. Our results showed that µG impairs the differentiation of osteocytes, consistent with prior osteoblast spaceflight experiments, which resulted in the downregulation of key osteocytic genes. Importantly, we demonstrate the modulation of critical glycolysis pathways in osteocytes subjected to microgravity and discovered a set of mechanical sensitive genes that are consistently regulated in multiple cell types exposed to microgravity suggesting a common, yet to be fully elucidated, genome-wide response to microgravity. Ground-based simulated microgravity experiments utilizing the NASA rotating-wall-vessel were unable to adequately replicate the changes in microgravity exposure highlighting the importance of spaceflight missions to understand the unique environmental stress that microgravity presents to diverse cell types. In summary, our findings demonstrate that osteocytes respond to µG with an increase in glucose metabolism and oxygen consumption.


Asunto(s)
Regulación de la Expresión Génica , Glucosa/metabolismo , Osteocitos/metabolismo , Consumo de Oxígeno , Vuelo Espacial/métodos , Transcriptoma , Animales , Mecanotransducción Celular , Ratones , Osteocitos/citología
2.
JCI Insight ; 4(5)2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843886

RESUMEN

During endochondral bone formation, chondrocyte hypertrophy represents a crucial turning point from chondrocyte differentiation to bone formation. Both parathyroid hormone-related protein (PTHrP) and histone deacetylase 4 (HDAC4) inhibit chondrocyte hypertrophy. Using multiple mouse genetics models, we demonstrate in vivo that HDAC4 is required for the effects of PTHrP on chondrocyte differentiation. We further show in vivo that PTHrP leads to reduced HDAC4 phosphorylation at the 14-3-3-binding sites and subsequent HDAC4 nuclear translocation. The Hdac4-KO mouse shares a similar but milder phenotype with the Pthrp-KO mouse, indicating the possible existence of other mediators of PTHrP action. We identify HDAC5 as an additional mediator of PTHrP signaling. While the Hdac5-KO mouse has no growth plate phenotype at birth, the KO of Hdac5 in addition to the KO of Hdac4 is required to block fully PTHrP action on chondrocyte differentiation at birth in vivo. Finally, we show that PTHrP suppresses myocyte enhancer factor 2 (Mef2) action that allows runt-related transcription factor 2 (Runx2) mRNA expression needed for chondrocyte hypertrophy. Our results demonstrate that PTHrP inhibits chondrocyte hypertrophy and subsequent bone formation in vivo by allowing HDAC4 and HDAC5 to block the Mef2/Runx2 signaling cascade. These results explain the phenotypes of several genetic abnormalities in humans.


Asunto(s)
Condrocitos/metabolismo , Histona Desacetilasas/metabolismo , Hipertrofia/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Animales , Cartílago/patología , Proliferación Celular , Condrocitos/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Histona Desacetilasas/genética , Humanos , Hipertrofia/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis/genética , Osteogénesis/fisiología , Proteína Relacionada con la Hormona Paratiroidea/genética , Fenotipo , Fosforilación , ARN Mensajero/metabolismo , Costillas/patología , Transducción de Señal , Transcriptoma
3.
Bone ; 117: 138-148, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30266511

RESUMEN

The stimulatory subunit of G-protein, Gsα, acts as a secondary messenger of G-protein coupled receptors (GPCRs) that primarily activates cAMP-induced signaling. GPCRs, such as the parathyroid hormone receptor (PTHR), are critical regulators of bone formation as shown by number of genetic manipulation studies targeting early osteoblast lineage cells. In this study, we have examined the role of Gsα in osteocytes, the terminally differentiated and most abundant cells of the osteoblast lineage. Mice lacking the stimulatory subunit of G-proteins (Gsα) in osteocytes (DMP1-GsαKO) have significant decrease of both trabecular and cortical bone, as assessed by µCT. Histomorphometric analysis showed that the osteopenia was mostly driven by more than 90% decrease in osteoblast numbers and activity whereas osteoclasts were only slightly decreased. The decrease in osteoblast number was associated with a striking lack of endocortical osteoblasts. We have previously shown that loss of the stimulatory subunit of G-proteins (Gsα) in osteocytes in vitro or in vivo induces high expression of sclerostin. To determine if the increased sclerostin levels contributed to the decreased endosteal bone lining cells and osteopenia, we treated wild-type mice with recombinant sclerostin and the DMP1-GsαKO mice with anti-sclerostin antibody. Treatment of wild-type mice with 100 µg/kg sclerostin for 3-weeks significantly reduced the numbers of bone lining cells and led to osteopenia. Next, the DMP1-GsαKO and control littermates were treated with the anti-sclerostin antibody (25 mg/kg, 2 times per week) for 4-weeks. Upon the antibody treatment, the endocortical osteoblasts reappeared in the DMP1-GsαKO mice to a comparable level to that of the vehicle treated control littermates. In control mice, E11/gp38 positive osteocytes were observed in parallel with the endocortical osteoblasts with higher dendrite density towards the endocortical osteoblasts. In DMP1-GsαKO mice, E11/gp38 positive osteocytes were lacking dendrites and were randomly scattered throughout the bone matrix. After treatment with anti-sclerostin antibody, DMP1-GsαKO mice showed increased E11/gp38 positive osteocytes near the endosteal bone surface and endosteal osteoblasts. The anti-sclerostin antibody treatment proportionally increased the bone volume but it could not completely rescue the osteopenia in the DMP1-GsαKO mice. Taken together, this data suggests that Gsα signaling in osteocytes leads to osteopenia driven, at least in part, by increased secretion of sclerostin.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Subunidades alfa de la Proteína de Unión al GTP Gs/deficiencia , Glicoproteínas/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Osteocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Anticuerpos Neutralizantes/metabolismo , Enfermedades Óseas Metabólicas/diagnóstico por imagen , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/metabolismo , Hueso Cortical/diagnóstico por imagen , Hueso Cortical/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Fémur/diagnóstico por imagen , Fémur/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis , Microtomografía por Rayos X
4.
J Bone Miner Res ; 32(2): 373-384, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27653320

RESUMEN

Cells of the osteoblast lineage are increasingly identified as participants in whole-body metabolism by primarily targeting pancreatic insulin secretion or consuming energy. Osteocytes, the most abundant bone cells, secrete a Wnt-signaling inhibitor called sclerostin. Here we examined three mouse models expressing high sclerostin levels, achieved through constitutive or inducible loss of the stimulatory subunit of G-proteins (Gsα in mature osteoblasts and/or osteocytes). These mice showed progressive loss of white adipose tissue (WAT) with tendency toward increased energy expenditure but no changes in glucose or insulin metabolism. Interestingly beige adipocytes were increased extensively in both gonadal and inguinal WAT and had reduced canonical ß-catenin signaling. To determine if sclerostin directly contributes to the increased beige adipogenesis, we engineered an osteocytic cell line lacking Gsα which has high sclerostin secretion. Conditioned media from these cells significantly increased expression of UCP1 in primary adipocytes, and this effect was partially reduced after depletion of sclerostin from the conditioned media. Similarly, treatment of Gsα-deficient animals with sclerostin-neutralizing antibody partially reduced the increased UCP1 expression in WAT. Moreover, direct treatment of sclerostin to wild-type mice significantly increased UCP1 expression in WAT. These results show that osteocytes and/or osteoblasts secrete factors regulating beige adipogenesis, at least in part, through the Wnt-signaling inhibitor sclerostin. Further studies are needed to assess metabolic effects of sclerostin on adipocytes and other metabolic tissues. © 2016 American Society for Bone and Mineral Research.


Asunto(s)
Adipogénesis , Tejido Adiposo Beige/metabolismo , Adiposidad , Glicoproteínas/metabolismo , Osteocitos/metabolismo , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales , Tejido Adiposo Blanco , Animales , Animales Recién Nacidos , Linaje de la Célula , Metabolismo Energético , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Ratones Noqueados , Tamaño de los Órganos , Osteoblastos/metabolismo , Fenotipo , Delgadez/metabolismo
5.
J Bone Miner Res ; 32(5): 892-901, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27862326

RESUMEN

Sclerostin antibody (Scl-Ab) increases osteoblast activity, in part through increasing modeling-based bone formation on previously quiescent surfaces. Histomorphometric studies have suggested that this might occur through conversion of bone lining cells into active osteoblasts. However, direct data demonstrating Scl-Ab-induced conversion of lining cells into active osteoblasts are lacking. Here, we used in vivo lineage tracing to determine if Scl-Ab promotes the conversion of lining cells into osteoblasts on periosteal and endocortical bone surfaces in mice. Two independent, tamoxifen-inducible lineage-tracing strategies were used to label mature osteoblasts and their progeny using the DMP1 and osteocalcin promoters. After a prolonged "chase" period, the majority of labeled cells on bone surfaces assumed a thin, quiescent morphology. Then, mice were treated with either vehicle or Scl-Ab (25 mg/kg) twice over the course of the subsequent week. After euthanization, marked cells were enumerated, their thickness quantified, and proliferation and apoptosis examined. Scl-Ab led to a significant increase in the average thickness of labeled cells on periosteal and endocortical bone surfaces, consistent with osteoblast activation. Scl-Ab did not induce proliferation of labeled cells, and Scl-Ab did not regulate apoptosis of labeled cells. Therefore, direct reactivation of quiescent bone lining cells contributes to the acute increase in osteoblast numbers after Scl-Ab treatment in mice. © 2016 American Society for Bone and Mineral Research.


Asunto(s)
Anticuerpos/farmacología , Hueso Cortical/metabolismo , Glicoproteínas/antagonistas & inhibidores , Osteoblastos/metabolismo , Periostio/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Hueso Cortical/citología , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Transgénicos , Osteoblastos/citología , Periostio/citología
6.
J Biol Chem ; 290(27): 16744-58, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25953900

RESUMEN

Although bone responds to its mechanical environment, the cellular and molecular mechanisms underlying the response of the skeleton to mechanical unloading are not completely understood. Osteocytes are the most abundant but least understood cells in bones and are thought to be responsible for sensing stresses and strains in bone. Sclerostin, a product of the SOST gene, is produced postnatally primarily by osteocytes and is a negative regulator of bone formation. Recent studies show that SOST is mechanically regulated at both the mRNA and protein levels. During prolonged bed rest and immobilization, circulating sclerostin increases both in humans and in animal models, and its increase is associated with a decrease in parathyroid hormone. To investigate whether SOST/sclerostin up-regulation in mechanical unloading is a cell-autonomous response or a hormonal response to decreased parathyroid hormone levels, we subjected osteocytes to an in vitro unloading environment achieved by the NASA rotating wall vessel system. To perform these studies, we generated a novel osteocytic cell line (Ocy454) that produces high levels of SOST/sclerostin at early time points and in the absence of differentiation factors. Importantly, these osteocytes recapitulated the in vivo response to mechanical unloading with increased expression of SOST (3.4 ± 1.9-fold, p < 0.001), sclerostin (4.7 ± 0.1-fold, p < 0.001), and the receptor activator of nuclear factor κΒ ligand (RANKL)/osteoprotegerin (OPG) (2.5 ± 0.7-fold, p < 0.001) ratio. These data demonstrate for the first time a cell-autonomous increase in SOST/sclerostin and RANKL/OPG ratio in the setting of unloading. Thus, targeted osteocyte therapies could hold promise as novel osteoporosis and disuse-induced bone loss treatments by directly modulating the mechanosensing cells in bone.


Asunto(s)
Glicoproteínas/genética , Osteocitos/metabolismo , Regulación hacia Arriba , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Fenómenos Biomecánicos , Línea Celular , Glicoproteínas/metabolismo , Gravitación , Péptidos y Proteínas de Señalización Intercelular , Ratones , Osteocitos/química , Ligando RANK/genética , Ligando RANK/metabolismo , Proteínas Wnt/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...