Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
J Dent Res ; 100(12): 1359-1366, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33899571

RESUMEN

Bone loss caused by trauma, neoplasia, congenital defects, or periodontal disease is a major cause of disability and human suffering. Skeletal progenitor cell-extracellular matrix interactions are critical for bone regeneration. Discoidin domain receptor 2 (DDR2), an understudied collagen receptor, plays an important role in skeletal development. Ddr2 loss-of-function mutations in humans and mice cause severe craniofacial and skeletal defects, including altered cranial shape, dwarfing, reduced trabecular and cortical bone, alveolar bone/periodontal defects, and altered dentition. However, the role of this collagen receptor in craniofacial regeneration has not been examined. To address this, calvarial subcritical-size defects were generated in wild-type (WT) and Ddr2-deficient mice. The complete bridging seen in WT controls at 4 wk postsurgery was not observed in Ddr2-deficient mice even after 12 wk. Quantitation of defect bone area by micro-computed tomography also revealed a 50% reduction in new bone volume in Ddr2-deficient mice. Ddr2 expression during calvarial bone regeneration was measured using Ddr2-LacZ knock-in mice. Expression was restricted to periosteal surfaces of uninjured calvarial bone and, after injury, was detected in select regions of the defect site by 3 d postsurgery and expanded during the healing process. The impaired bone healing associated with Ddr2 deficiency may be related to reduced osteoprogenitor or osteoblast cell proliferation and differentiation since knockdown/knockout of Ddr2 in a mesenchymal cell line and primary calvarial osteoblast cultures reduced osteoblast differentiation while Ddr2 overexpression was stimulatory. In conclusion, Ddr2 is required for cranial bone regeneration and may be a novel target for therapy.


Asunto(s)
Regeneración Ósea , Receptor con Dominio Discoidina 2 , Cráneo , Animales , Ratones , Osteoblastos , Microtomografía por Rayos X
2.
J Dent Res ; 99(2): 214-222, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31869264

RESUMEN

Collagen signaling is critical for proper bone and tooth formation. Discoidin domain receptor 2 (DDR2) is a collagen-activated tyrosine kinase receptor shown to be essential for skeletal development. Patients with loss of function mutations in DDR2 develop spondylo-meta-epiphyseal dysplasia (SMED), a rare, autosomal recessive disorder characterized by short stature, short limbs, and craniofacial anomalies. A similar phenotype was observed in Ddr2-deficient mice, which exhibit dwarfism and defective bone formation in the axial, appendicular, and cranial skeletons. However, it is not known if Ddr2 has a role in tooth formation. We first defined the expression pattern of Ddr2 during tooth formation using Ddr2-LacZ knock-in mice. Ddr2 expression was detected in the dental follicle/sac and dental papilla mesenchyme of developing teeth and in odontoblasts and the periodontal ligament (PDL) of adults. No LacZ staining was detected in wild-type littermates. This Ddr2 expression pattern suggests a potential role in the tooth and surrounding periodontium. To uncover the function of Ddr2, we used Ddr2slie/slie mice, which contain a spontaneous 150-kb deletion in the Ddr2 locus to produce an effective null. In comparison with wild-type littermates, Ddr2slie/slie mice displayed disproportional tooth size (decreased root/crown ratio), delayed tooth root development, widened PDL space, and interradicular alveolar bone defects. Ddr2slie/slie mice also had abnormal collagen content associated with upregulation of periostin levels within the PDL. The delayed root formation and periodontal abnormalities may be related to defects in RUNX2-dependent differentiation of odontoblasts and osteoblasts; RUNX2-S319-P was reduced in PDLs from Ddr2slie/slie mice, and deletion of Ddr2 in primary cell cultures from dental pulp and PDL inhibited differentiation of cells to odontoblasts or osteoblasts, respectively. Together, our studies demonstrate odontoblast- and PDL-specific expression of Ddr2 in mature and immature teeth, as well as indicate that DDR2 signaling is important for normal tooth formation and maintenance of the surrounding periodontium.


Asunto(s)
Receptor con Dominio Discoidina 2 , Odontogénesis , Animales , Receptor con Dominio Discoidina 2/genética , Receptores con Dominio Discoidina , Humanos , Ratones , Odontogénesis/genética , Proteínas Tirosina Quinasas Receptoras , Receptores Mitogénicos/genética
3.
J Dent Res ; 87(9): 845-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18719211

RESUMEN

BMP2/7 heterodimer expression by adenovirus can stimulate bone formation at subcutaneous sites. In the present study, we evaluate whether this approach will also promote healing of cranial defects. Adenovirus expressing BMP2 or BMP7 (AdBMP2, AdBMP7) was titrated to yield equivalent BMP protein levels after transduction into murine BLK cells. Analysis of conditioned medium showed that BMP2/7 heterodimers have enhanced ability to stimulate alkaline phosphatase and Smad 1,5,8 phosphorylation relative to equivalent amounts of BMP2 or BMP7 homodimers. To measure bone regeneration, we implanted virally transduced BLK cells into critical-sized calvarial defects generated in C57BL6 mice. AdBMP2/7-transduced cells were more effective in healing cranial defects than were cells individually transduced with AdBMP2 or BMP7. Dramatic increases in bone volume fraction, as measured by microCT, as well as fusion of regenerated bone with the defect margins were noted. Thus, the use of gene therapy to express heterodimeric BMPs is a promising potential therapy for healing craniofacial bones.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Regeneración Ósea/fisiología , Terapia Genética/métodos , Regeneración Tisular Dirigida/métodos , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/fisiología , Implantes Absorbibles , Adenoviridae/genética , Animales , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/administración & dosificación , Proteínas Morfogenéticas Óseas/genética , Regeneración Ósea/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Craneotomía , Fibroblastos/citología , Fibroblastos/metabolismo , Esponja de Gelatina Absorbible/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Ratones , Ratones Endogámicos C57BL , Mioblastos/citología , Mioblastos/metabolismo , Osteogénesis/genética , Osteogénesis/fisiología , Andamios del Tejido , Factor de Crecimiento Transformador beta/administración & dosificación , Factor de Crecimiento Transformador beta/genética , Transgenes , Cicatrización de Heridas/genética
4.
J Dent Res ; 97(3): 321-328, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29073363

RESUMEN

Temporomandibular joint (TMJ) disorders are often associated with development of osteoarthritis-like changes in the mandibular condyle. Discoidin domain receptor 2 (DDR2), a collagen receptor preferentially activated by type I and III collagen found in the TMJ and other fibrocartilages, has been associated with TMJ degeneration, but its role in normal joint development has not been previously examined. Using Ddr2 LacZ-tagged mice and immunohistochemistry, we found that DDR2 is preferentially expressed and activated in the articular zone of TMJs but not knee joints. To assess the requirement for Ddr2 in TMJ development, studies were undertaken to compare wild-type and smallie ( slie) mice, which contain a spontaneous deletion in Ddr2 to produce an effective null allele. Analysis of TMJs from newborn Ddr2slie/slie mice revealed a developmental delay in condyle mineralization, as measured by micro-computed tomography and histologic analysis. In marked contrast, knee joints of Ddr2slie/slie mice were normal. Analysis of older Ddr2slie/slie mice (3 and 10 mo) revealed that the early developmental delay led to a dramatic and progressive loss of TMJ articular integrity and osteoarthritis-like changes. Mutant condyles had a rough and flattened bone surface, accompanied by a dramatic loss of bone mineral density. Mankin scores showed significantly greater degenerative changes in the TMJs of 3- and 10-mo-old Ddr2slie/slie mice as compared with wild-type controls. No DDR2-dependent degenerative changes were seen in knees. Analysis of primary cultures of TMJ articular chondrocytes from wild-type and Ddr2slie/slie mice showed defects in chondrocyte maturation and mineralization in the absence of Ddr2. These studies demonstrate that DDR2 is necessary for normal TMJ condyle development and homeostasis and that these DDR2 functions are restricted to TMJ fibrocartilage and not seen in the hyaline cartilage of the knee.


Asunto(s)
Envejecimiento/fisiología , Receptor con Dominio Discoidina 2/fisiología , Articulación de la Rodilla/crecimiento & desarrollo , Articulación Temporomandibular/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Cartílago Articular/crecimiento & desarrollo , Diferenciación Celular , Condrocitos/fisiología , Inmunohistoquímica , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Coloración y Etiquetado , Articulación Temporomandibular/diagnóstico por imagen , Microtomografía por Rayos X
5.
J Dent Res ; 86(12): 1207-11, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18037657

RESUMEN

Marrow stromal cells (MSCs) include stem cells capable of forming all mesenchymal tissues, including bone. However, before MSCs can be successfully used in regeneration procedures, methods must be developed to stimulate their differentiation selectively to osteoblasts. Runx2, a bone-specific transcription factor, is known to stimulate osteoblast differentiation. In the present study, we tested the hypothesis that Runx2 gene therapy can be used to heal a critical-sized defect in mouse calvaria. Runx2-engineered MSCs displayed enhanced osteogenic potential and osteoblast-specific gene expression in vitro and in vivo. Runx2-expressing cells also dramatically enhanced the healing of critical-sized calvarial defects and increased both bone volume fraction and bone mineral density. These studies provide a novel route for enhancing osteogenesis that may have future therapeutic applications for craniofacial bone regeneration.


Asunto(s)
Células de la Médula Ósea/citología , Regeneración Ósea/genética , Diferenciación Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Osteogénesis/genética , Trasplante de Células Madre , Animales , Células de la Médula Ósea/metabolismo , Células Cultivadas , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Cráneo , Células del Estroma/trasplante , Transducción Genética , Cicatrización de Heridas/genética
6.
Oncogene ; 35(3): 366-76, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25867060

RESUMEN

The osteogenic transcription factor, Runx2, is abnormally expressed in prostate cancer (PCa) and associated with metastatic disease. During bone development, Runx2 is activated by signals known to be hyperactive in PCa including the RAS/MAP kinase pathway, which phosphorylates Runx2 on multiple serine residues including S301 and S319 (equivalent to S294 and S312 in human Runx2). This study examines the role of these phosphorylation sites in PCa. Runx2 was preferentially expressed in more invasive PCa cell lines (PC3>C4-2B>LNCaP). Furthermore, analysis using a P-S319-Runx2-specific antibody revealed that the ratio of P-S319-Runx2/total Runx2 as well as P-ERK/total ERK was highest in PC3 followed by C4-2B and LNCaP cells. These results were confirmed by immunofluorescence confocal microscopy, which showed a higher percentage of PC3 cells staining positive for P-S319-Runx2 relative to C4-2B and LNCaP cells. Phosphorylated Runx2 had an exclusively nuclear localization. When expressed in prostate cell lines, wild-type Runx2 increased metastasis-associated gene expression, in vitro migratory and invasive activity as well as in vivo growth of tumor cell xenografts. In contrast, S301A/S319A phosphorylation site mutations greatly attenuated these Runx2 responses. Analysis of tissue microarrays from 129 patients revealed strong nuclear staining with the P-S319-Runx2 antibody in primary PCas and metastases. P-S319-Runx2 staining was positively correlated with Gleason score and occurrence of lymph node metastases while little or no Runx2 phosphorylation was seen in normal prostate, benign prostate hyperplasia or prostatitis indicating that Runx2 S319 phosphorylation is closely associated with PCa induction and progression towards an aggressive phenotype. These studies establish the importance of Runx2 phosphorylation in prostate tumor growth and highlight its value as a potential diagnostic marker and therapeutic target.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Fosforilación/genética , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Dent Res ; 84(12): 1093-103, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16304438

RESUMEN

Safe, effective approaches for bone regeneration are needed to reverse bone loss caused by trauma, disease, and tumor resection. Unfortunately, the science of bone regeneration is still in its infancy, with all current or emerging therapies having serious limitations. Unlike current regenerative therapies that use single regenerative factors, the natural processes of bone formation and repair require the coordinated expression of many molecules, including growth factors, bone morphogenetic proteins, and specific transcription factors. As will be developed in this article, future advances in bone regeneration will likely incorporate therapies that mimic critical aspects of these natural biological processes, using the tools of gene therapy and tissue engineering. This review will summarize current knowledge related to normal bone development and fracture repair, and will describe how gene therapy, in combination with tissue engineering, may mimic critical aspects of these natural processes. Current gene therapy approaches for bone regeneration will then be summarized, including recent work where combinatorial gene therapy was used to express groups of molecules that synergistically interacted to stimulate bone regeneration. Last, proposed future directions for this field will be discussed, where regulated gene expression systems will be combined with cells seeded in precise three-dimensional configurations on synthetic scaffolds to control both temporal and spatial distribution of regenerative factors. It is the premise of this article that such approaches will eventually allow us to achieve the ultimate goal of bone tissue engineering: to reconstruct entire bones with associated joints, ligaments, or sutures. Abbreviations used: BMP, bone morphogenetic protein; FGF, fibroblast growth factor; AER, apical ectodermal ridge; ZPA, zone of polarizing activity; PZ, progress zone; SHH, sonic hedgehog; OSX, osterix transcription factor; FGFR, fibroblast growth factor receptor; PMN, polymorphonuclear neutrophil; PDGF, platelet-derived growth factor; IGF, insulin-like growth factor; TGF-beta, tumor-derived growth factor beta; CAR, coxsackievirus and adenovirus receptor; MLV, murine leukemia virus; HIV, human immunodeficiency virus; AAV, adeno-associated virus; CAT, computer-aided tomography; CMV, cytomegalovirus; GAM, gene-activated matrix; MSC, marrow stromal cell; MDSC, muscle-derived stem cell; VEGF, vascular endothelial growth factor.


Asunto(s)
Regeneración Ósea/fisiología , Terapia Genética , Biomimética , Regeneración Ósea/genética , Humanos , Ingeniería de Tejidos
8.
Mol Endocrinol ; 11(8): 1103-13, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9212058

RESUMEN

Osteocalcin is a hormonally regulated calcium-binding protein made almost exclusively by osteoblasts. In normal cells, osteocalcin expression requires ascorbic acid (AA), an essential cofactor for osteoblast differentiation both in vivo and in vitro. To determine the mechanism of this regulation, subclones of MC3T3-E1 preosteoblasts were transiently transfected with 1.3 kb of the mouse osteocalcin gene 2 promoter driving expression of firefly luciferase. AA stimulated luciferase activity 20-fold after 4-5 days. This response was stereospecific to L-ascorbic acid and was only detected in MC3T3-E1 subclones showing strong AA induction of the endogenous osteocalcin gene. Similar results were also obtained in MC3T3-E1 cells stably transfected with the osteocalcin promoter. A specific inhibitor of collagen synthesis, 3,4-dehydroproline, blocked AA-dependent induction of promoter activity, indicating that regulation of the osteocalcin gene requires collagen matrix synthesis. Deletion analysis of the mOG2 promoter identified an essential region for AA responsiveness between -147 and -116 bp. This region contains a single copy of the previously described osteoblast-specific element, OSE2. Deletion and mutation of OSE2 in DNA transfection assays established the requirement for this element in the AA response. Furthermore, DNA-binding assays revealed that MC3T3-E1 cells contain OSF2, the nuclear factor binding to OSE2, and that binding of OSF2 to OSE2 is up-regulated by AA treatment. Taken collectively, our results indicate that an intact OSE2 sequence is required for the induction of osteocalcin expression by AA.


Asunto(s)
Ácido Ascórbico/metabolismo , Moléculas de Adhesión Celular/metabolismo , Colágeno/biosíntesis , Osteoblastos/metabolismo , Osteocalcina/genética , Animales , Ácido Ascórbico/farmacología , Sitios de Unión , Células Cultivadas , Colágeno/efectos de los fármacos , Ratones , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteocalcina/efectos de los fármacos , Osteocalcina/metabolismo , Prolina/análogos & derivados , Prolina/farmacología , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Tiempo , Transfección
9.
Bone ; 71: 244-56, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25460184

RESUMEN

Bone cell culture systems are essential tools for the study of the molecular mechanisms regulating extracellular matrix mineralization. MC3T3-E1 osteoblast cell cultures are the most commonly used in vitro model of bone matrix mineralization. Despite the widespread use of this cell line to study biomineralization, there is as yet no systematic characterization of the mineral phase produced in these cultures. Here we provide a comprehensive, multi-technique biophysical characterization of this cell culture mineral and extracellular matrix, and compare it to mouse bone and synthetic apatite mineral standards, to determine the suitability of MC3T3-E1 cultures for biomineralization studies. Elemental compositional analysis by energy-dispersive X-ray spectroscopy (EDS) showed calcium and phosphorus, and trace amounts of sodium and magnesium, in both biological samples. X-ray diffraction (XRD) on resin-embedded intact cultures demonstrated that similar to 1-month-old mouse bone, apatite crystals grew with preferential orientations along the (100), (101) and (111) mineral planes indicative of guided biogenic growth as opposed to dystrophic calcification. XRD of crystals isolated from the cultures revealed that the mineral phase was poorly crystalline hydroxyapatite with 10 to 20nm-sized nanocrystallites. Consistent with the XRD observations, electron diffraction patterns indicated that culture mineral had low crystallinity typical of biological apatites. Fourier-transform infrared spectroscopy (FTIR) confirmed apatitic carbonate and phosphate within the biological samples. With all techniques utilized, cell culture mineral and mouse bone mineral were remarkably similar. Scanning (SEM) and transmission (TEM) electron microscopy showed that the cultures had a dense fibrillar collagen matrix with small, 100nm-sized, collagen fibril-associated mineralization foci which coalesced to form larger mineral aggregates, and where mineralized sites showed the accumulation of the mineral-binding protein osteopontin. Light microscopy, confocal microscopy and three-dimensional reconstructions showed that some cells had dendritic processes and became embedded within the mineral in an osteocyte-like manner. In conclusion, we have documented characteristics of the mineral and matrix phases of MC3T3-E1 osteoblast cultures, and have determined that the structural and compositional properties of the mineral are highly similar to that of mouse bone.


Asunto(s)
Huesos/fisiología , Huesos/ultraestructura , Calcificación Fisiológica , Matriz Extracelular/metabolismo , Osteoblastos/fisiología , Osteoblastos/ultraestructura , Animales , Células Cultivadas , Ratones , Minerales/metabolismo , Espectrometría por Rayos X , Espectroscopía Infrarroja por Transformada de Fourier , Vibración , Difracción de Rayos X
10.
J Bone Miner Res ; 7(2): 235-46, 1992 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1373931

RESUMEN

The MC3T3-E1 mouse calvaria-derived cell line has been used to study the role of collagen synthesis in osteoblast differentiation. MC3T3-E1 cells, like several previously characterized osteoblast culture systems, expressed osteoblast markers and formed a mineralized extracellular matrix only after exposure to ascorbic acid. Mineralization was stimulated further by beta-glycerol phosphate. Ultrastructural observations indicated that the extracellular matrix produced by ascorbic acid-treated cells was highly organized and contained well-banded collagen fibrils. Expression of osteoblast markers followed a clear temporal sequence. The earliest effects of ascorbic acid were to stimulate type I procollagen mRNA and collagen synthesis (24 h after ascorbate addition), followed by induction of alkaline phosphatase (48-72 h) and osteocalcin (96-144 h) mRNAs. Procollagen mRNA, which was expressed constitutively in the absence of ascorbate, increased only twofold after vitamin C addition. In contrast, alkaline phosphatase and osteocalcin mRNAs were undetectable in untreated cultures. Actions of ascorbic acid on osteoblast marker gene expression are mediated by increases in collagen synthesis and/or accumulation because (1) parallel dose-response relationships were obtained for ascorbic acid stimulation of collagen accumulation and alkaline phosphatase activity, and (2) the specific collagen synthesis inhibitors, 3,4-dehydroproline and cis-4-hydroxyproline, reversibly blocked ascorbic acid-dependent collagen synthesis and osteoblast marker gene expression.


Asunto(s)
Ácido Ascórbico/fisiología , Colágeno/biosíntesis , Matriz Extracelular/fisiología , Osteoblastos/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/química , Diferenciación Celular/fisiología , Línea Celular , Colágeno/análisis , Ratones , Microscopía Electrónica , Minerales/metabolismo , Fenotipo , ARN/aislamiento & purificación , ARN Mensajero/biosíntesis , Difracción de Rayos X
11.
J Bone Miner Res ; 5(11): 1157-67, 1990 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1702922

RESUMEN

The bone, liver, and kidney isozyme of alkaline phosphatase (ALP) has been measured in MG-63 human osteosarcoma cells after treatment with ascorbic acid (AA) and/or 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3]. Both compounds were required to achieve maximum ALP activity. When grown in the absence of 1,25-(OH)2D3 cells had low basal ALP activity regardless of whether media contained AA. In AA-free medium, 1,25-(OH)2D3 (10 nM) increased ALP activity fourfold. Addition of AA further increased levels of ALP activity induced by 1,25-(OH)2D3 to 10-15 times those found in -AA controls. The earliest effects of 1,25-(OH)2D3 were seen after 24-48 h, and ALP activity continued to increase for 6-8 days. AA and 1,25-(OH)2D3 had similar effects on ALP activity in ROS 17/2.8 rat osteosarcoma cells. In MG-63 cells the effects of AA and 1,25-(OH)2D3 could not be simply explained by the ability of these compounds to inhibit cell growth because another mitotic inhibitor, hydroxyurea, had a minimal effect on ALP activity. 1,25-(OH)2D3-specific induction of ALP +/- AA was totally blocked by inhibitors of protein and RNA synthesis. Maximal ALP induction was obtained when cells were plated at low density. Consistent with our previous report (Franceschi et al. 1988 J Biol Chem 263:18938-18945), 1,25-(OH)2D3 rapidly stimulated type I collagen synthesis and acid-precipitable hydroxyproline production in MG-63 cells and this stimulation was further increased by AA. These results suggest that induction of the osteoblast marker, ALP, is directly or indirectly coupled to collagen matrix synthesis and/or accumulation.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Ácido Ascórbico/farmacología , Huesos/efectos de los fármacos , Calcitriol/farmacología , Huesos/metabolismo , Recuento de Células , División Celular/efectos de los fármacos , Línea Celular , Colágeno/biosíntesis , Glicerofosfatos/farmacología , Humanos , Isoenzimas/metabolismo , Cinética , Biosíntesis de Proteínas , ARN/biosíntesis
12.
J Bone Miner Res ; 9(6): 843-54, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8079660

RESUMEN

Treatment of mouse MC3T3-E1 cells with ascorbic acid initiates the formation of a collagenous extracellular matrix and synthesis of several osteoblast-related proteins. We recently showed that ascorbic acid dramatically increases alkaline phosphatase and osteocalcin mRNAs and that this induction is blocked by inhibitors of collagen triple-helix formation (Franceschi and Iyer, J Bone Miner Res 7:235). In the present study, the relationship between collagen matrix formation and osteoblast-specific gene expression is explored in greater detail. Kinetic studies revealed that ascorbic acid increased proline hydroxylation in the intracellular procollagen pool within 1 h and stimulated the cleavage of type I collagen propeptides beginning at 2.5 h. Mature alpha 1(I) and alpha 2(I) collagen components were first detected at 10 h and continued to increase in both cell layer and culture medium for up to 72 h. Ascorbic acid also increased the rate of procollagen secretion from cell layers to culture medium. The secretion of another matrix protein, fibronectin, was only slightly affected. Alkaline phosphatase or its mRNA was first detected 2-3 days after ascorbic acid addition, but osteocalcin mRNA was not seen until day 6. Two inhibitors of collagen triple-helix formation, ethyl-3,4-dihydroxybenzoate and 3,4-dehydroproline, inhibited procollagen hydroxylation and alkaline phosphatase induction. 3,4-Dehydroproline also inhibited the induction of alkaline phosphatase and osteocalcin mRNAs. Surprisingly, induction was not blocked if cells were exposed to ascorbic acid before inhibitor addition. Alkaline phosphatase was also partially inhibited if cells were grown in the presence of purified bacterial collagenase. These results indicate that the induction of osteoblast markers by ascorbic acid does not require the continuous hydroxylation and processing of procollagens and suggest that a stable, possibly matrix-associated signal is generated at early times after ascorbic acid addition that allows subsequent induction of osteoblast-related genes.


Asunto(s)
Ácido Ascórbico/farmacología , Colágeno/biosíntesis , Matriz Extracelular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Células 3T3 , Fosfatasa Alcalina/biosíntesis , Animales , Diferenciación Celular/efectos de los fármacos , Colagenasas/farmacología , Inducción Enzimática/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Expresión Génica , Hidroxilación , Ratones , Osteoblastos/citología , Osteocalcina/biosíntesis , Procolágeno/metabolismo , Prolina/metabolismo , ARN Mensajero/metabolismo
13.
J Bone Miner Res ; 10(12): 1969-77, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8619378

RESUMEN

A well-defined chicken osteoblast culture system(18) has been used to examine fibronectin (FN) mRNA levels, synthesis, and accumulation during in vitro differentiation and matrix mineralization. Immunofluorescent staining of cells after 6 or 18 days in culture revealed that FN was initially associated with the cell surface and in partial coalignment with cytoskeletal elements while at the latter time most FN was associated with the extracellular matrix as a ubiquitous fibrillar network. Western blot analysis of total cell-associated proteins also detected FN at all culture times. However, when results were normalized to cellular DNA, FN levels increased until 12-16 and remained relatively constant thereafter. Similarly, FN synthesis as measured by [35S]-methionine labeling, and immunoprecipitation was greatest in early cultures (culture day 3) and then declined such that synthesis decreased 60% at day 18 and 94% after 24-31 days. FN mRNA levels as measured by Northern blot analysis were well correlated with FN synthesis. These results clearly show that FN is made by primary osteoblasts during their in vitro maturation. In contrast to other osteoblast markers such as alkaline phosphatase, osteocalcin, and osteopontin, whose expression increases as cells differentiate, FN accumulates in the matrix during periods of early cell growth and attachment and then remains proportional to cell number. Results with FN differ from those obtained with collagen which continues to accumulate in the extracellular matrix during osteoblast maturation. These results are consistent with FN being important for the initial attachment of early osteoblasts or osteoblast precursors to the pericellular matrix.


Asunto(s)
Fibronectinas/genética , Regulación del Desarrollo de la Expresión Génica/genética , Osteoblastos/citología , Actinas/metabolismo , Animales , Autorradiografía , Western Blotting , Adhesión Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Embrión de Pollo , ADN/metabolismo , Fibronectinas/biosíntesis , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente , ARN Mensajero/metabolismo
14.
J Bone Miner Res ; 10(11): 1635-43, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8592939

RESUMEN

When grown in medium containing ascorbic acid and beta-glycerol phosphate, mouse MC3T3-E1 cells express an osteoblast phenotype and produce a highly mineralized extracellular matrix. The purpose of this study was to independently examine the role of the collagenous matrix and functional osteoblasts on the mineralization process. Cultures with and without an extensive collagenous matrix were prepared by growing MC3T3-E1 cells in the presence and absence of ascorbic acid. Matrix-rich cultures mineralized at much lower calcium phosphate ion products than nonmatrix cultures. At higher ion products, spontaneous precipitation in the medium and cell layers of nonmatrix cultures were observed. In contrast, mineral in matrix-rich cultures was still exclusively associated with collagen fibrils and not with ectopic sites in the cell layer or medium. To examine the effect of cell viability on matrix mineralization, cells were grown 8 or 16 days in the presence of ascorbic acid, then killed and incubated in a mineralizing medium. Significant mineralization was not observed in the collagenous matrix of 8-day killed cultures or age-matched controls. At 16 days mineral was associated with collagen fibrils at specific foci in the matrix of both viable and killed cultures. This observation is consistent with the concept that collagenous matrices must undergo a maturation process before they can support a mineral induction and growth. It further shows that osteoblast-like cells are not required for mineralization of mature matrices, but are required for matrix maturation.


Asunto(s)
Calcificación Fisiológica/fisiología , Matriz Extracelular/fisiología , Osteoblastos/fisiología , Células 3T3 , Animales , Ácido Ascórbico/farmacología , Calcificación Fisiológica/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Glicerofosfatos/farmacología , Ratones , Microscopía Electrónica , Osteoblastos/ultraestructura
15.
J Bone Miner Res ; 14(6): 893-903, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10352097

RESUMEN

A series of subclonal cell lines with high or low differentiation/mineralization potential after growth in the presence of ascorbic acid (AA) were derived from murine MC3T3-E1 cells. Subclones were characterized in terms of their ability to mineralize a collagenous extracellular matrix both in vitro and in vivo and express osteoblast-related genes. When compared with nonmineralizing cells, mineralizing subclones selectively expressed mRNAs for the osteoblast markers, bone sialoprotein (BSP), osteocalcin (OCN), and the parathyroid hormone (PTH)/parathyroid hormone-related protein (PTHrP) receptor. In contrast, alkaline phosphatase mRNA was present in certain nonmineralizing as well as mineralizing subclones, suggesting that its expression may be subject to different controls from other osteoblast markers. Only highly differentiating subclones exhibited strong AA-dependent induction of a transiently transfected OCN promoter-luciferase reporter gene, indicating that there was a good correlation between mRNA levels and transcriptional activity. Consistent with its postulated role in biomineralization, BSP as measured by Western blotting was only present in mineralizing subclones. After implantation into immunodeficient mice, highly differentiating subclones formed bone-like ossicles resembling woven bone, while poorly differentiating cells only produced fibrous tissue. Interestingly, subclones with both high and low differentiation potential produced similar amounts of collagen in culture and expressed comparable basal levels of mRNA encoding Osf2/Cbfa1, an osteoblast-related transcription factor. Although some strongly differentiating cells exhibited a modest AA-dependent up-regulation of Osf2/Cbfa1 mRNA, there was no clear relationship between levels of this message and induction of mRNAs for other differentiation markers. Thus, the mere presence of Osf2/Cbfa1 in a subclone was not sufficient for osteoblast differentiation. These subclones will be very useful for studying critical events in osteoblast differentiation and mineralization.


Asunto(s)
Densidad Ósea/fisiología , Osteoblastos/citología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Células Clonales , Regulación de la Expresión Génica/fisiología , Ratones , Regiones Promotoras Genéticas
16.
J Bone Miner Res ; 14(3): 396-405, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10027904

RESUMEN

Bone sialoprotein (BSP) is an extracellular matrix protein that is intimately associated with the process of biomineralization. Osf2, a member of the Cbf/runt family of transcription factors, is required for the development of osteoblasts in vivo and has been reported to stimulate the transcription of BSP when overexpressed in mesenchymal cell lines. To investigate the role of Osf2 in BSP expression, we cloned a 2.5 kb fragment of a 5' untranscribed sequence from the murine BSP gene and evaluated it for putative Osf2 binding sites. This promoter, which was able to direct 5- to 10-fold higher levels of luciferase reporter expression in osteoblastic cells than in nonbone cell lines, contains two consensus core binding sites for members of the Cbf/runt family. One, at -61 relative to the start of transcription, is within a region having 75% overall sequence identity with the rat and human BSP promoters. The other is located at -1335, outside this highly conserved region. Neither site is completely conserved in the rat or human sequences. Only the -1335 site was able to bind a protein in nuclear extracts of osteoblastic cells, and this protein was identified as Osf2. Despite this in vitro binding ability, we detected no significant enhancer activity in the -1335 element when placed in front of a minimal osteocalcin promoter driving a luciferase reporter gene in osteoblastic cells nor any loss in transcriptional activity of a 5' promoter deletion which eliminated this element as compared with the full-length 2.5 kb promoter. These results suggest that Osf2 binding to the BSP promoter is not essential for its osteoblast-selective expression.


Asunto(s)
Proteínas de Neoplasias , Regiones Promotoras Genéticas , Sialoglicoproteínas/genética , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Secuencia de Bases , Sitios de Unión/genética , Clonación Molecular , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Factores de Unión al Sitio Principal , ADN/genética , ADN/metabolismo , Cartilla de ADN/genética , Elementos de Facilitación Genéticos , Humanos , Sialoproteína de Unión a Integrina , Ratones , Datos de Secuencia Molecular , Mutación , Ratas , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie
17.
J Bone Miner Res ; 10(8): 1243-55, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8585429

RESUMEN

TGF beta has opposing effects on osteoblasts which are thought to be differentiation stage dependent; however, little is known concerning the effects of TGF beta on osteoblastic characteristics at different stages of maturation. The purpose of this study was to characterize the pattern of mRNA expression for the PTH/PTHrP receptor during normal osteoblastic differentiation in vitro, and evaluate the effects of TGF beta 1 on PTH/PTHrP receptor and osteocalcin (OCN) steady-state mRNA at different stages of osteoblastic differentiation. MC3T3-E1 preosteoblasts were plated at low density and induced to differentiate with ascorbic acid and beta-glycerophosphate. The first group served as a vehicle control and the remaining five groups received a single 48 h TGF beta 1 (3.0 ng/ml)-pulse staggered on a weekly basis for 30 days. Cell cultures were harvested weekly and evaluated for: steady-state PTH/PTHrP receptor and OCN mRNA levels via northern analysis, calcium and phosphorous levels, bone nodules via Von Kossa staining, alkaline phosphatase enzyme levels, and hydroxyproline levels. Group 1 (control) samples followed a normal pattern of proliferation, extracellular matrix deposition, and mineralization. PTH/PTHrP receptor and OCN mRNA expression increased 8-fold and 10-fold respectively, over the collection periods. When TGF beta 1 was administered during the first 48 h period (group 2) while cells were rapidly proliferating, there was a persistent inhibition of PTH/PTHrP receptor expression and a striking reduction in OCN mRNA expression at all time points. There was also a down-regulation of PTH/PTHrP receptor and OCN expression when TGF beta 1 was administered later during osteoblast differentiation (groups 3-6); however, these effects were not persistent. In addition there was a total lack of bone nodule formation in group two cultures, whereas groups 3-6 had increasing bone nodule formation because the TGF beta 1 was administered later in the culture period. These studies indicate that expression of the PTH/PTHrP receptor increases with osteoblastic differentiation and suggest that TGF beta 1 inhibits osteoblastic maturation with more persistent effects found in less differentiated osteoblastic cells.


Asunto(s)
Osteoblastos/efectos de los fármacos , Osteocalcina/genética , ARN Mensajero/metabolismo , Receptores de Hormona Paratiroidea/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Células 3T3/efectos de los fármacos , Células 3T3/metabolismo , Animales , Ácido Ascórbico/farmacología , Northern Blotting , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Glicerofosfatos/farmacología , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/metabolismo
18.
Hum Gene Ther ; 11(8): 1201-10, 2000 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-10834621

RESUMEN

An ex vivo gene therapy strategy was used to achieve localized skeletal regeneration in vivo. When an adenovirus vector engineered to express bone morphogenetic protein 7 transduced human gingival fibroblasts or rat dermal fibroblasts, these nonosteogenic tissues formed bone and supported the development of hematopoietic tissue when transplanted into immunocompromised mice. Transduced gingival fibroblasts formed marrow-containing ossicles in 100% of transplants after 1-2 weeks in vivo (n = 30). Immunostaining with murine and human-specific antisera raised against osteonectin and in situ hybridization of human-specific Alu genomic sequence demonstrated that the newly formed bone organ was a chimera of both the human donor and the mouse recipient cells. In experiments of greater clinical relevance, AdCMVBMP-7-transduced dermal fibroblasts repaired critical size skeletal defects in rat calvariae (n = 6). The results of this study suggest a bifunctional role of BMP-7-transduced fibroblasts. The transduced, nonosteogenic cells not only secreted biologically active BMP-7 in vitro and in vivo, but also differentiated into bone-forming cells in vivo. This model exploits the use of an easily biopsied, self-regenerating tissue such as gingiva or skin and suggests that local regeneration of tissues by ex vivo gene therapy may not require that autogenous cells be cultured from the tissue that is to be regenerated.


Asunto(s)
Desarrollo Óseo/genética , Proteínas Morfogenéticas Óseas/genética , Terapia Genética/métodos , Osteogénesis/genética , Transducción Genética , Factor de Crecimiento Transformador beta , Adenoviridae/genética , Elementos Alu/genética , Animales , Western Blotting , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/biosíntesis , Craneotomía , Fibroblastos/metabolismo , Encía/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Osteonectina/metabolismo , Ratas , Ratas Endogámicas Lew , Piel/metabolismo , Cráneo/metabolismo
19.
Endocrinology ; 141(12): 4671-80, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11108282

RESUMEN

PTH-related protein (PTHrP) acts as a paracrine and/or autocrine regulator of cell proliferation, apoptosis, and differentiation and is implicated in tooth development. The current studies employed cementoblasts to determine the role(s) and mechanisms of PTHrP in regulating cementum formation. Results demonstrated that PTHrP repressed gene expression and protein synthesis of bone sialoprotein (BSP) and abolished cementoblast-mediated biomineralization in vitro. The BSP gene inhibition required protein synthesis. The PTHrP analog (1-31) and other activators of the PKA pathway (3-isobutyl-1-methylxathine (IBMX), forskolin (FSK) and Sp-Adenosine-3', 5'-cyclic monophosphorothioate (Sp-cAMPss) also down-regulated BSP gene expression and blocked cementoblast-mediated biomineralization. In contrast, the PTHrP analog (7-34), a PTHrP antagonist, and the activators of the PKC pathway [phorbol 12-myristate 13-acetate (PMA) and phorbol 12, 13-dibutyrate (PDBu)] promoted BSP gene expression. In addition, the PKA pathway inhibitor (9-(2-tetrahydrofuryl) adenine (THFA) partially, but significantly reversed the PTHrP-mediated down-regulation of BSP gene expression. Furthermore, THFA alone significantly increased BSP messenger RNA (mRNA) expression in cementoblasts. In contrast, the inhibitor of the PKC pathway (GF109203X) did not reverse the PTHrP inhibitory effect on BSP gene expression. Furthermore, GF109203X alone dramatically reduced the BSP transcript levels. These data indicate that the cAMP/PKA pathway mediates the PTHrP-mediated down-regulation of BSP mRNA expression in cementoblasts; and furthermore, this pathway may, through an intrinsic inhibition mechanism, regulate the basal level of BSP mRNA expression. In contrast, the activation of PKC promotes BSP gene expression. These data provide new insights into the molecular mechanisms involved in PTHrP regulation of cementogenesis.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/análogos & derivados , Cemento Dental/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas/farmacología , Sialoglicoproteínas/genética , 1-Metil-3-Isobutilxantina/farmacología , Línea Celular , Colforsina/farmacología , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Cicloheximida/farmacología , Dactinomicina/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Sialoproteína de Unión a Integrina , Cinética , Proteína Relacionada con la Hormona Paratiroidea , Forbol 12,13-Dibutirato/farmacología , Proteínas/administración & dosificación , ARN Mensajero/análisis , Sistemas de Mensajero Secundario , Acetato de Tetradecanoilforbol/farmacología , Tionucleótidos/farmacología
20.
Endocrinology ; 142(10): 4379-88, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11564701

RESUMEN

As part of its overall function as a major regulator of calcium homeostasis, PTH stimulates bone resorption and inhibits osteoblast-mediated biomineralization. To determine the basis for the inhibitory actions of this hormone, we compared the time course of PTH-dependent inhibition of mineralization in MC3T3-E1 osteoblast-like cells with changes in mRNA levels for several extracellular matrix proteins previously associated either with induction or inhibition of mineralization. Mineralizing activity was rapidly lost in PTH-treated cells ( approximately 30% inhibition after 3 h, 50% inhibition at 6 h). Of the proteins examined, changes in matrix gamma-carboxyglutamic acid protein were best correlated with PTH-dependent inhibition of mineralization. Matrix gamma-carboxyglutamic acid protein mRNA was rapidly induced 3 h after PTH treatment, with a 6- to 8-fold induction seen after 6 h. Local in vivo injection of PTH over the calvaria of mice also induced a 2-fold increase in matrix gamma-carboxyglutamic acid protein mRNA. Warfarin, an inhibitor of matrix gamma-carboxyglutamic acid protein gamma-carboxylation, reversed the effects of PTH on mineralization in MC3T3-E1 cells, whereas vitamin K enhanced PTH activity, as would be expected if a gamma-carboxyglutamic acid-containing protein were required for PTH activity. Levels of the other mRNAs examined were not well correlated with the observed changes in mineralization. Osteopontin, an in vitro inhibitor of mineralization, was induced approximately 4-fold 12 h after PTH addition. Bone sialoprotein mRNA, which encodes an extracellular matrix component most frequently associated with mineral induction, was inhibited by 50% after 12 h of PTH treatment. Osteocalcin mRNA, encoding the other known gamma-carboxyglutamic acid protein in bone, was also inhibited by PTH, but, again, with a significantly slower time course than was seen for mineral inhibition. Taken together, these results show that the rapid inhibition of osteoblast mineralization induced by in vitro PTH treatment is at least in part explained by induction of matrix gamma-carboxyglutamic acid protein.


Asunto(s)
Proteínas de la Matriz Extracelular/fisiología , Osteoblastos/fisiología , Hormona Paratiroidea/fisiología , Ácido 1-Carboxiglutámico/metabolismo , Animales , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/fisiología , Línea Celular , Ratones , Hormona Paratiroidea/farmacología , ARN Mensajero/análisis , Sialoglicoproteínas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA