Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Calcif Tissue Int ; 109(1): 66-76, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33616712

RESUMEN

Osteocyte produced fibroblast growth factor 23 (FGF23) is the key regulator of serum phosphate (Pi) homeostasis. The interplay between parathyroid hormone (PTH), FGF23 and other proteins that regulate FGF23 production and serum Pi levels is complex and incompletely characterised. Evidence suggests that the protein product of the SOST gene, sclerostin (SCL), also a PTH target and also produced by osteocytes, plays a role in FGF23 expression, however the mechanism for this effect is unclear. Part of the problem of understanding the interplay of these mediators is the complex multi-organ system that achieves Pi homeostasis in vivo. In the current study, we sought to address this using a cell line model of the osteocyte, IDG-SW3, known to express FGF23 at both the mRNA and protein levels. In cultures of differentiated IDG-SW3 cells, both PTH1-34 and recombinant human (rh) SCL remarkably induced Fgf23 mRNA expression dose-dependently within 3 h. Both rhPTH1-34 and rhSCL also strongly induced C-terminal FGF23 protein secretion. Secreted intact FGF23 levels remained unchanged, consistent with constitutive post-translational cleavage of FGF23 in this cell model. Both rhPTH1-34 and rhSCL treatments significantly suppressed mRNA levels of Phex, Dmp1 and Enpp1 mRNA, encoding putative negative regulators of FGF23 levels, and induced Galnt3 mRNA expression, encoding N-acetylgalactosaminyl-transferase 3 (GalNAc-T3), which protects FGF23 from furin-like proprotein convertase-mediated cleavage. The effect of both rhPTH1-34 and rhSCL was antagonised by pre-treatment with the NF-κß signalling inhibitors, BAY11 and TPCK. RhSCL also stimulated FGF23 mRNA expression in ex vivo cultures of human bone. These findings provide evidence for the direct regulation of FGF23 expression by sclerostin. Locally expressed sclerostin via the induction of FGF23 in osteocytes thus has the potential to contribute to the regulation of Pi homeostasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Crecimiento de Fibroblastos , Osteocitos , Animales , Huesos , Diferenciación Celular , Factor-23 de Crecimiento de Fibroblastos , Humanos , Ratones
2.
Calcif Tissue Int ; 95(2): 183-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24916279

RESUMEN

The central importance of osteocytes in regulating bone homeostasis is becoming increasingly apparent. However, the study of these cells has been restricted by the relative paucity of cell line models, especially those of human origin. Therefore, we investigated the extent to which SaOS2 human osteosarcoma cells can differentiate into osteocyte-like cells. During culture under the appropriate mineralising conditions, SaOS2 cells reproducibly synthesised a bone-like mineralised matrix and temporally expressed the mature osteocyte marker genes SOST, DMP1, PHEX and MEPE and down-regulated expression of RUNX2 and COL1A1. SaOS2 cells cultured in 3D collagen gels acquired a dendritic morphology, characteristic of osteocytes, with multiple interconnecting cell processes. These findings suggest that SaOS2 cells have the capacity to differentiate into mature osteocyte-like cells under mineralising conditions. PTH treatment of SaOS2 cells resulted in strong down-regulation of SOST mRNA expression at all time points tested. Interestingly, PTH treatment resulted in the up-regulation of RANKL mRNA expression only at earlier stages of differentiation. These findings suggest that the response to PTH is dependent on the differentiation stage of the osteoblast/osteocyte. Together, our results demonstrate that SaOS2 cells can be used as a human model to investigate responses to osteotropic stimuli throughout differentiation to a mature osteocyte-like stage.


Asunto(s)
Calcificación Fisiológica/fisiología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Osteoblastos/citología , Osteocitos/citología , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral/citología , Humanos , Técnicas In Vitro , Microscopía Confocal , Osteoblastos/metabolismo , Osteocitos/metabolismo , Osteosarcoma , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría por Rayos X
3.
ACS Biomater Sci Eng ; 7(2): 441-450, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33492936

RESUMEN

There is an increasing demand for low-cost and more efficient titanium (Ti) medical implants that will provide improved osseointegration and at the same time reduce the likelihood of infection. In the past decade, additive manufacturing (AM) using metal selective laser melting (SLM) or three-dimensional (3D) printing techniques has emerged to enable novel implant geometries or properties to overcome such potential challenges. This study presents a new surface engineering approach to create bioinspired multistructured surfaces on SLM-printed Ti alloy (Ti6Al4V) implants by combining SLM technology, electrochemical anodization, and hydrothermal (HT) processes. The resulting implants display unique surfaces with a distinctive dual micro- to nano-topography composed of micron-sized spherical features, fabricated by SLM and vertically aligned nanoscale pillar structures as a result of combining anodization and HT treatment. The fabricated implants enhanced hydroxyapatite-like mineral deposition from simulated body fluid (SBF) compared to control. In addition, normal human osteoblast-like cells (NHBCs) showed strong adhesion to the nano-/microstructures and displayed greater propensity to mineralize compared to control surfaces. This engineering approach and the resulting nature-inspired multiscale-structured surface offers desired features for improving osseointegration and antibacterial performance toward the development of next-generation orthopedic and dental implants.


Asunto(s)
Prótesis e Implantes , Titanio , Humanos , Ensayo de Materiales , Oseointegración , Propiedades de Superficie
4.
Sci Rep ; 11(1): 22593, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799645

RESUMEN

Mesenchymal progenitors differentiate into several tissues including bone, cartilage, and adipose. Targeting these cells in vivo is challenging, making mesenchymal progenitor cell lines valuable tools to study tissue development. Mesenchymal stem cells (MSCs) can be isolated from humans and animals; however, obtaining homogenous, responsive cells in a reproducible fashion is challenging. As such, we developed two mesenchymal progenitor cell (MPC) lines, MPC1 and MPC2, generated from bone marrow of male C57BL/6 mice. These cells were immortalized using the temperature sensitive large T-antigen, allowing for thermal control of proliferation and differentiation. Both MPC1 and MPC2 cells are capable of osteogenic, adipogenic, and chondrogenic differentiation. Under osteogenic conditions, both lines formed mineralized nodules, and stained for alizarin red and alkaline phosphatase, while expressing osteogenic genes including Sost, Fgf23, and Dmp1. Sost and Dmp1 mRNA levels were drastically reduced with addition of parathyroid hormone, thus recapitulating in vivo responses. MPC cells secreted intact (iFGF23) and C-terminal (cFGF23) forms of the endocrine hormone FGF23, which was upregulated by 1,25 dihydroxy vitamin D (1,25D). Both lines also rapidly entered the adipogenic lineage, expressing adipose markers after 4 days in adipogenic media. MPC cells were also capable of chondrogenic differentiation, displaying increased expression of cartilaginous genes including aggrecan, Sox9, and Comp. With the ability to differentiate into multiple mesenchymal lineages and mimic in vivo responses of key regulatory genes/proteins, MPC cells are a valuable model to study factors that regulate mesenchymal lineage allocation as well as the mechanisms that dictate transcription, protein modification, and secretion of these factors.


Asunto(s)
Adipocitos/citología , Técnicas de Cultivo de Célula , Condrocitos/citología , Células Madre Mesenquimatosas/citología , Osteocitos/citología , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Proliferación Celular , Factor-23 de Crecimiento de Fibroblastos/metabolismo , Inmunofenotipificación , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
5.
Am J Physiol Cell Physiol ; 297(6): C1358-67, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19675304

RESUMEN

The vitamin K family members phylloquinone (vitamin K1) and the menaquinones (vitamin K2) are under study for their roles in bone metabolism and as potential therapeutic agents for skeletal diseases. We have investigated the effects of two naturally occurring homologs, phytonadione (vitamin K1) and menatetrenone (vitamin K2), and those of the synthetic vitamin K, menadione (vitamin K3), on human primary osteoblasts. All homologs promoted in vitro mineralization by these cells. Vitamin K1-induced mineralization was highly sensitive to warfarin, whereas that induced by vitamins K2 and K3 was less sensitive, implying that gamma-carboxylation and other mechanisms, possibly genomic actions through activation of the steroid xenobiotic receptor, are involved in the effect. The positive effect on mineralization was associated with decreased matrix synthesis, evidenced by a decrease from control in expression of type I collagen mRNA, implying a maturational effect. Incubation in the presence of vitamin K2 or K3 in a three-dimensional type I collagen gel culture system resulted in increased numbers of cells with elongated cytoplasmic processes resembling osteocytes. This effect was not warfarin sensitive. Addition of calcein to vitamin K-treated cells revealed vitamin K-dependent deposition of mineral associated with cell processes. These effects are consistent with vitamin K promoting the osteoblast-to-osteocyte transition in humans. To test whether vitamin K may also act on mature osteocytes, we tested the effects of vitamin K on MLO-Y4 cells. Vitamin K reduced receptor activator of NF-kappaB ligand expression relative to osteoprotegerin by MLO-Y4 cells, an effect also seen in human cultures. Together, our findings suggest that vitamin K promotes the osteoblast-to-osteocyte transition, at the same time decreasing the osteoclastogenic potential of these cells. These may be mechanisms by which vitamin K optimizes bone formation and integrity in vivo and may help explain the net positive effect of vitamin K on bone formation.


Asunto(s)
Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas/metabolismo , Osteoblastos/citología , Osteocitos/citología , Vitamina K/farmacología , Ácido 1-Carboxiglutámico/metabolismo , Proliferación Celular/efectos de los fármacos , Humanos , Metabolismo/efectos de los fármacos , Fenotipo , Vitamina K 1/farmacología , Vitamina K 2/farmacología , Vitamina K 3/farmacología , Warfarina/farmacología
6.
J Steroid Biochem Mol Biol ; 177: 83-90, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28887147

RESUMEN

Previous studies have shown that 1α,25-dihydroxyvitamin D3 (1,25D) through vitamin D receptor (VDR) signalling has both catabolic and anabolic effects on osteoblast differentiation. However, the mechanism of these differential effects by 1,25D is not fully understood. In this study, mice with three different genetic backgrounds, representing a normal VDR level (wild-type, WT), VDR over-expression specifically in mature osteoblasts (ObVDR-B6) and global VDR knockout (VDRKO), were utilised to generate primary osteoblast-like cultures to further elucidate the effects of 1,25D on osteoblast differentiation. Our data confirm the importance of VDR in the late stage of osteogenic differentiation and also for the expression of factors critical for osteoblastic support of osteoclast formation. This study also demonstrates the differential effects of a pharmacological level of 1,25D (1nM) on the expression of osteogenic differentiation markers, including Ocn and Sost, depending on the relative level of VDR. Our findings suggest that 1,25D plays an inhibitory role in matrix mineralisation, possibly through the modulation of the tissue non-specific alkaline phosphatase to ectonucleotide pyrophosphatase/phosphodiesterase 1 axis, in a VDR level-dependent manner. We conclude that the relative VDR level and the 1,25D availability to cells, are important co-determinants for whether 1,25D plays a promoting or suppressive role in osteoblast-mediated osteogenic activity.


Asunto(s)
Osteoblastos/efectos de los fármacos , Receptores de Calcitriol/fisiología , Vitamina D/análogos & derivados , Proteínas Adaptadoras Transductoras de Señales , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular , Ligandos , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/fisiología , Osteocalcina/genética , Vitamina D/farmacología
7.
mBio ; 9(2)2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29691335

RESUMEN

Periprosthetic joint infection (PJI) is a potentially devastating complication of orthopedic joint replacement surgery. PJI with associated osteomyelitis is particularly problematic and difficult to cure. Whether viable osteocytes, the predominant cell type in mineralized bone tissue, have a role in these infections is not clear, although their involvement might contribute to the difficulty in detecting and clearing PJI. Here, using Staphylococcus aureus, the most common pathogen in PJI, we demonstrate intracellular infection of human-osteocyte-like cells in vitro and S. aureus adaptation by forming quasi-dormant small-colony variants (SCVs). Consistent patterns of host gene expression were observed between in vitro-infected osteocyte-like cultures, an ex vivo human bone infection model, and bone samples obtained from PJI patients. Finally, we confirm S. aureus infection of osteocytes in clinical cases of PJI. Our findings are consistent with osteocyte infection being a feature of human PJI and suggest that this cell type may provide a reservoir for silent or persistent infection. We suggest that elucidating the molecular/cellular mechanism(s) of osteocyte-bacterium interactions will contribute to better understanding of PJI and osteomyelitis, improved pathogen detection, and treatment.IMPORTANCE Periprosthetic joint infections (PJIs) are increasing and are recognized as one of the most common modes of failure of joint replacements. Osteomyelitis arising from PJI is challenging to treat and difficult to cure and increases patient mortality 5-fold. Staphylococcus aureus is the most common pathogen causing PJI. PJI can have subtle symptoms and lie dormant or go undiagnosed for many years, suggesting persistent bacterial infection. Osteocytes, the major bone cell type, reside in bony caves and tunnels, the lacuno-canalicular system. We report here that S. aureus can infect and reside in human osteocytes without causing cell death both experimentally and in bone samples from patients with PJI. We demonstrate that osteocytes respond to infection by the differential regulation of a large number of genes. S. aureus adapts during intracellular infection of osteocytes by adopting the quasi-dormant small-colony variant (SCV) lifestyle, which might contribute to persistent or silent infection. Our findings shed new light on the etiology of PJI and osteomyelitis in general.


Asunto(s)
Osteoartritis/fisiopatología , Osteocitos/microbiología , Infecciones Relacionadas con Prótesis/fisiopatología , Infecciones Estafilocócicas/fisiopatología , Staphylococcus aureus/patogenicidad , Células Cultivadas , Humanos , Modelos Biológicos
8.
Bone ; 88: 64-72, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27109824

RESUMEN

Osteocytes are essential regulators of bone homeostasis. However, they are difficult to study due to their location within the bone mineralised matrix. Although several techniques have been published for the isolation of osteocytes from mouse bone, no such technique has been described for human osteocytes. We have therefore developed a protocol for the isolation of osteocytes from human trabecular bone samples acquired during surgery. The cells were digested from the bone matrix by sequential collagenase and ethylenediaminetetraacetic acid (EDTA) digestions and the cells from later digests displayed characteristic dendritic osteocyte morphology when cultured ex vivo. Furthermore, the cells expressed characteristic osteocyte marker genes, such as E11, dentin matrix protein 1 (DMP1), SOST, matrix extracellular phosphoglycoprotein (MEPE) and phosphate regulating endopeptidase homologue, X-linked (PHEX). In addition, genes associated with osteocyte perilacunar remodelling, including matrix metallopeptidase-13 (MMP13), cathepsin K (CTSK) and carbonic anhydrase 2 (CAR2) were expressed. The cells also responded to parathyroid hormone (PTH) by downregulating SOST mRNA expression and to 1α,25-dihydroxyvitamin D3 (1,25D) by upregulating fibroblast growth factor 23 (FGF23) mRNA expression. Therefore, the cells behave in a similar manner to osteocytes in vivo. These cells represent an important tool in enhancing current knowledge in human osteocyte biology.


Asunto(s)
Hueso Esponjoso/citología , Separación Celular/métodos , Osteocitos/citología , Animales , Biomarcadores/metabolismo , Remodelación Ósea , Forma de la Célula , Células Cultivadas , Células Endoteliales/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Regulación de la Expresión Génica , Hematopoyesis , Osteocitos/metabolismo , Donantes de Tejidos
9.
J Steroid Biochem Mol Biol ; 164: 369-373, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26690786

RESUMEN

The osteocyte expressed gene SOST encodes sclerostin, a potent negative regulator of bone formation and inducer of bone resorption. We have recently demonstrated that the human SOST gene is positively regulated in response to 1α,25-dihydroxyvitamin D3 (1,25D). Responsiveness may be mediated at least in part by a single classical DR3-type vitamin D response element (VDRE). In this study we examined the early responsiveness of the SOST gene to both 1,25D and to parathyroid hormone (PTH), a known repressor of SOST expression, in SaOS2 cells differentiated to an osteocyte-like stage of cell maturation. Both SOST mRNA levels and sclerostin protein levels increased in these cultures as early as 3h post-treatment with 1,25D and declined in response to PTH in the same timeframe. For 1,25D, the level of induced SOST appeared dependent on the extent, to which the degradative enzyme 1,25-dihydroxyvitamin D 24-hydroxylase (CYP24A1) was induced. Together with the observed rapid decrease in SOST/sclerostin levels in response to PTH, endocrine regulation of sclerostin production appears to be an important determinant of sclerostin levels. These findings confirm that the human SOST gene and sclerostin expression can be considered to be directly 1,25D-responsive in osteocytes.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Marcadores Genéticos/genética , Osteocitos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Vitamina D/análogos & derivados , Proteínas Adaptadoras Transductoras de Señales , Proteínas Morfogenéticas Óseas/agonistas , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Humanos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteocitos/citología , Osteocitos/metabolismo , Hormona Paratiroidea/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Transducción de Señal , Vitamina D/metabolismo , Vitamina D/farmacología , Vitamina D3 24-Hidroxilasa/genética , Vitamina D3 24-Hidroxilasa/metabolismo
10.
Mol Cell Endocrinol ; 399: 208-18, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25458698

RESUMEN

Fibroblast growth factor-23 (FGF23), produced by osteocytes, is the key physiological regulator of phosphate homeostasis. Sepsis patients often experience transient hypophosphataemia, suggesting the regulation of FGF23 levels by pro-inflammatory factors. Here, we used the osteocyte-like cell line IDG-SW3 to investigate the effect of pro-inflammatory stimuli on FGF23 production. In differentiated IDG-SW3 cultures, basal Fgf23 mRNA was dose-dependently up-regulated by pro-inflammatory cytokines TNF, IL-1ß and TWEAK, and bacterial LPS. Similar effects were observed in human bone samples. TNF- and IL-1ß-induced Fgf23 expression was NF-κB-dependent. Conversely, mRNA encoding negative regulators of FGF23, Phex, Dmp1 and Enpp1, were suppressed by TNF, IL-1ß, TWEAK and LPS, independent of NF-κß signalling. Galnt3, the protein product of which protects intact FGF23 protein from furin/furin-like proprotein convertase cleavage, increased in response to these treatments. C-terminal FGF23 and intact FGF23 protein levels also increased, the latter only in the presence of Furin inhibitors, suggesting that enzymatic cleavage exerts critical control of active FGF23 secretion by osteocytes. Our results demonstrate in principle that pro-inflammatory stimuli are capable of increasing osteocyte secretion of FGF23, which may contribute to hypophosphataemia during sepsis and possibly other inflammatory conditions.


Asunto(s)
Huesos/metabolismo , Diferenciación Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Osteocitos/metabolismo , Huesos/patología , Línea Celular , Citocina TWEAK , Proteínas de la Matriz Extracelular/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/toxicidad , Lipopolisacáridos/toxicidad , Osteocitos/patología , Endopeptidasa Neutra Reguladora de Fosfato PHEX/metabolismo , Fosfoproteínas/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Pirofosfatasas/metabolismo , Factor de Necrosis Tumoral alfa/toxicidad , Factores de Necrosis Tumoral/toxicidad
11.
Mol Cell Endocrinol ; 412: 140-7, 2015 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-26054750

RESUMEN

While vitamin D supplementation is common, the anabolic mechanisms that improve bone status are poorly understood. Under standard mineralising conditions including media ionised calcium of 1.1 mM, 1,25-dihydroxyvitamin D3 (1,25D) enhanced differentiation and mineral deposition by the mature osteoblast/pre-osteocyte cell line, MLO-A5. This effect was markedly increased with a higher ionised calcium level (1.5 mM). Gene expression analyses revealed that 1,25D-induced mineral deposition was associated with induction of Enpp1 mRNA, coding for nucleotide pyrophosphatase phosphodiesterase 1 (NPP1) and NPP1 protein levels. Since MLO-A5 cells express abundant alkaline phosphatase that was not further modified by 1,25D treatment or exposure to increased calcium, this finding suggested that the NPP1 production of pyrophosphate (PPi) may provide alkaline phosphatase with substrate for the generation of inorganic phosphate (Pi). Consistent with this, co-treatment with Enpp1 siRNA or a NPP1 inhibitor, PPADS, abrogated 1,25D-induced mineral deposition. These data demonstrate that 1,25D stimulates osteoblast differentiation and mineral deposition, and interacts with the extracellular calcium concentration. 1,25D regulates Enpp1 expression, which presumably, in the context of adequate tissue non-specific alkaline phosphatase activity, provides Pi to stimulate mineralisation. Our findings suggest a mechanism by which vitamin D with adequate dietary calcium can improve bone mineral status.


Asunto(s)
Calcio/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Pirofosfatasas/metabolismo , Vitamina D/análogos & derivados , Animales , Calcificación Fisiológica , Diferenciación Celular , Línea Celular , Expresión Génica , Ratones , Osteoblastos , Osteocalcina/genética , Osteocalcina/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Pirofosfatasas/genética , Receptores de Calcitriol/metabolismo , Vitamina D/farmacología
12.
Mol Cell Endocrinol ; 413: 157-67, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26112182

RESUMEN

Sclerostin, the SOST gene product, is a negative regulator of bone formation and a positive regulator of bone resorption. In this study, treatment of human primary osteoblasts, including cells differentiated to an osteocyte-like stage, with 1α,25-dihydroxyvitaminD3 (1,25D) resulted in the dose-dependent increased expression of SOST mRNA. A similar effect was observed in human trabecular bone samples cultured ex vivo, and in osteocyte-like cultures of differentiated SAOS2 cells. Treatment of SAOS2 cells with 1,25D resulted in the production and secretion of sclerostin protein. In silico analysis of the human SOST gene revealed a single putative DR3-type vitamin D response element (VDRE) at position -6216 bp upstream of the transcription start site (TSS). This sequence was confirmed to have strong VDRE activity by luciferase reporter assays and electrophoretic mobility shift analysis (EMSA). Sequence substitution in the VDR/RXR half-sites abolished VDRE reporter activity and binding of nuclear proteins. A 6.3 kb fragment of the human proximal SOST promoter demonstrated responsiveness to 1,25D. The addition of the evolutionary conserved region 5 (ECR5), a known bone specific enhancer region, ahead of the 6.3 kb fragment increased basal promoter activity but did not increase 1,25D responsiveness. Site-specific mutagenesis abolished the responsiveness of the 6.3 kb promoter to 1,25D. We conclude that 1,25D is a direct regulator of human SOST gene and sclerostin protein expression, extending the pathways of control of sclerostin expression. At least some of this responsiveness is mediated by the identified classical VDRE however the nature of the transcriptional regulation by 1,25D warrants further investigation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Calcitriol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Osteocitos/metabolismo , Transcripción Genética/efectos de los fármacos , Elemento de Respuesta a la Vitamina D/fisiología , Proteínas Adaptadoras Transductoras de Señales , Proteínas Morfogenéticas Óseas/genética , Línea Celular Tumoral , Regulación de la Expresión Génica/fisiología , Marcadores Genéticos/genética , Humanos , Osteocitos/citología , Transcripción Genética/fisiología
13.
J Bone Miner Res ; 28(12): 2436-48, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23737439

RESUMEN

The osteocyte product sclerostin is emerging as an important paracrine regulator of bone mass. It has recently been shown that osteocyte production of receptor activator of NF-κB ligand (RANKL) is important in osteoclastic bone resorption, and we reported that exogenous treatment of osteocytes with sclerostin can increase RANKL-mediated osteoclast activity. There is good evidence that osteocytes can themselves liberate mineral from bone in a process known as osteocytic osteolysis. In the current study, we investigated sclerostin-stimulated mineral dissolution by human primary osteocyte-like cells (hOCy) and mouse MLO-Y4 cells. We found that sclerostin upregulated osteocyte expression of carbonic anhydrase 2 (CA2/Car2), cathepsin K (CTSK/Ctsk), and tartrate-resistant acid phosphatase (ACP5/Acp5). Because acidification of the extracellular matrix is a critical step in the release of mineral from bone, we further examined the regulation by sclerostin of CA2. Sclerostin stimulated CA2 mRNA and protein expression in hOCy and in MLO-Y4 cells. Sclerostin induced a decrease in intracellular pH (pHi) in both cell types as well as a decrease in extracellular pH (pHo) and the release of calcium ions from mineralized substrate. These effects were reversed in the co-presence of the carbonic anhydrase inhibitor, acetozolamide. Car2-siRNA knockdown in MLO-Y4 cells significantly inhibited the ability of sclerostin to both reduce the pHo and release calcium from a mineralized substrate. Knockdown in MLO-Y4 cells of each of the putative sclerostin receptors, Lrp4, Lrp5 and Lrp6, using siRNA, inhibited the sclerostin induction of Car2, Catk and Acp5 mRNA, as well as pHo and calcium release. Consistent with this activity of sclerostin resulting in osteocytic osteolysis, human trabecular bone samples treated ex vivo with recombinant human sclerostin for 7 days exhibited an increased osteocyte lacunar area, an effect that was reversed by the co-addition of acetozolamide. These findings suggest a new role for sclerostin in the regulation of perilacunar mineral by osteocytes.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Huesos/metabolismo , Anhidrasas Carbónicas/biosíntesis , Glicoproteínas/metabolismo , Minerales/metabolismo , Osteocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Huesos/efectos de los fármacos , Calcio/metabolismo , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Marcadores Genéticos , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Osteocitos/efectos de los fármacos , Osteocitos/enzimología , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/farmacología
14.
PLoS One ; 6(10): e25900, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21991382

RESUMEN

Sclerostin is a product of mature osteocytes embedded in mineralised bone and is a negative regulator of bone mass and osteoblast differentiation. While evidence suggests that sclerostin has an anti-anabolic role, the possibility also exists that sclerostin has catabolic activity. To test this we treated human primary pre-osteocyte cultures, cells we have found are exquisitely sensitive to sclerostin, or mouse osteocyte-like MLO-Y4 cells, with recombinant human sclerostin (rhSCL) and measured effects on pro-catabolic gene expression. Sclerostin dose-dependently up-regulated the expression of receptor activator of nuclear factor kappa B (RANKL) mRNA and down-regulated that of osteoprotegerin (OPG) mRNA, causing an increase in the RANK:OPG mRNA ratio. To examine the effects of rhSCL on resulting osteoclastic activity, MLO-Y4 cells plated onto a bone-like substrate were primed with rhSCL for 3 days and then either mouse splenocytes or human peripheral blood mononuclear cells (PBMC) were added. This resulted in cultures with elevated osteoclastic resorption (approximately 7-fold) compared to untreated co-cultures. The increased resorption was abolished by co-addition of recombinant OPG. In co-cultures of MLO-Y4 cells with PBMC, SCL also increased the number and size of the TRAP-positive multinucleated cells formed. Importantly, rhSCL had no effect on TRAP-positive cell formation from monocultures of either splenocytes or PBMC. Further, rhSCL did not induce apoptosis of MLO-Y4 cells, as determined by caspase activity assays, demonstrating that the osteoclastic response was not driven by dying osteocytes. Together, these results suggest that sclerostin may have a catabolic action through promotion of osteoclast formation and activity by osteocytes, in a RANKL-dependent manner.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Osteoclastos/citología , Osteocitos/citología , Ligando RANK/metabolismo , Transducción de Señal/efectos de los fármacos , Fosfatasa Ácida/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adulto , Animales , Apoptosis/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Humanos , Isoenzimas/metabolismo , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Ligando RANK/genética , Proteínas Recombinantes/farmacología , Fosfatasa Ácida Tartratorresistente
15.
J Bone Miner Res ; 26(7): 1425-36, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21312267

RESUMEN

The identity of the cell type responsive to sclerostin, a negative regulator of bone mass, is unknown. Since sclerostin is expressed in vivo by mineral-embedded osteocytes, we tested the hypothesis that sclerostin would regulate the behavior of cells actively involved in mineralization in adult bone, the preosteocyte. Differentiating cultures of human primary osteoblasts exposed to recombinant human sclerostin (rhSCL) for 35 days displayed dose- and time-dependent inhibition of in vitro mineralization, with late cultures being most responsive in terms of mineralization and gene expression. Treatment of advanced (day 35) cultures with rhSCL markedly increased the expression of the preosteocyte marker E11 and decreased the expression of mature markers DMP1 and SOST. Concomitantly, matrix extracellular phosphoglycoprotein (MEPE) expression was increased by rhSCL at both the mRNA and protein levels, whereas PHEX was decreased, implying regulation through the MEPE-ASARM axis. We confirmed that mineralization by human osteoblasts is exquisitely sensitive to the triphosphorylated ASARM-PO4 peptide. Immunostaining revealed that rhSCL increased the endogenous levels of MEPE-ASARM. Importantly, antibody-mediated neutralization of endogenous MEPE-ASARM antagonized the effect of rhSCL on mineralization, as did the PHEX synthetic peptide SPR4. Finally, we found elevated Sost mRNA expression in the long bones of HYP mice, suggesting that sclerostin may drive the increased MEPE-ASARM levels and mineralization defect in this genotype. Our results suggest that sclerostin acts through regulation of the PHEX/MEPE axis at the preosteocyte stage and serves as a master regulator of physiologic bone mineralization, consistent with its localization in vivo and its established role in the inhibition of bone formation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Calcificación Fisiológica , Diferenciación Celular , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Osteoblastos/citología , Osteocitos/citología , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Anticuerpos Neutralizantes/farmacología , Proteínas Morfogenéticas Óseas/farmacología , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Humanos , Hipofosfatemia/metabolismo , Hipofosfatemia/patología , Péptidos y Proteínas de Señalización Intercelular , Ratones , Modelos Biológicos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Endopeptidasa Neutra Reguladora de Fosfato PHEX/metabolismo , Proteínas Recombinantes/farmacología
16.
J Bone Miner Res ; 24(8): 1434-49, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19292615

RESUMEN

We have recently shown that TNF-related weak inducer of apoptosis (TWEAK) is a mediator of inflammatory bone remodeling. The aim of this study was to investigate the role of TWEAK in modulating human osteoblast activity, and how TWEAK and TNFalpha might interact in this context. Recombinant TWEAK and TNF were both mitogenic for human primary osteoblasts (NHBC). TWEAK dose- and time-dependently regulated the expression of the osteoblast transcription factors RUNX2 and osterix. TWEAK inhibited in vitro mineralization and downregulated the expression of osteogenesis-associated genes. Significantly, TWEAK and TWEAK/TNF induced the expression of the osteoblast differentiation inhibitor and SOST gene product, sclerostin. Sclerostin induction was mitogen-activated protein kinase (MAPK) dependent. The SOST mRNA levels induced by TWEAK were equivalent to or exceeded those seen in steady-state human bone, and the TWEAK/TNF induction of SOST mRNA was recapitulated in fresh cancellous bone explants. TWEAK-induced sclerostin expression was observed in immature osteoblastic cells, both in cycling (Ki67(+)) primary NHBC and in the cell lines MC3T3-E1 and MG-63, as well as in human osteocyte-like cells and in the osteocyte cell line, MLO-Y4. Treatment of NHBC with recombinant human sclerostin mimicked the effects of TWEAK to suppress RUNX2 and osteocalcin (OCN). TWEAK, TNF, and sclerostin treatment of NHBC similarly altered levels of phosphorylated and total GSK3beta and active and total levels of beta-catenin, implying that the Wnt signaling pathway was affected by all three stimuli. Sclerostin also rapidly activated ERK-1/2 MAPK signaling, indicating the involvement of additional signaling pathways. Together, our findings suggest that TWEAK, alone and with TNF, can regulate osteoblast function, at least in part by inducing sclerostin expression. Our results also suggest new roles and modes of action for sclerostin.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Mediadores de Inflamación/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoblastos/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factores de Necrosis Tumoral/farmacología , Células 3T3 , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Western Blotting , Proteínas Morfogenéticas Óseas/genética , Proliferación Celular/efectos de los fármacos , Citocina TWEAK , Cartilla de ADN , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Marcadores Genéticos/genética , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Osteoblastos/citología , Osteoblastos/enzimología , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA