Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Osteoporos ; 2023: 8998556, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36660551

RESUMEN

In previous study, we showed that nucleolar protein 66 (NO66) is a chromatin modifier and negatively regulates Osterix activity as well as mesenchymal progenitor differentiation. Genetic ablation of the NO66 (RIOX1) gene in cells of the Prx1-expressing mesenchymal lineage leads to acceleration of osteochondrogenic differentiation and a larger skeleton in adult mice, whereas mesenchyme-specific overexpression of NO66 inhibits osteochondrogenesis resulting in dwarfism and osteopenia. However, the impact of NO66 overexpression in cells of the osteoblast lineage in vivo remains largely undefined. Here, we generated osteoblast-specific transgenic mice overexpressing a FLAG-tagged NO66 transgene driven by the 2.3 kB alpha-1type I collagen (Col1a1) promoter. We found that overexpression of NO66 in cells of the osteoblast lineage did not cause overt defects in developmental bones but led to osteoporosis in the long bones of adult mice. This includes decreased bone volume (BV), bone volume density (bone volume/total volume, BV/TV), and bone mineral density (BMD) in cancellous compartment of long bones, along with the accumulation of fatty droplets in bone marrow. Ex vivo culture of the bone marrow mesenchymal stem/stromal cells (BMSCs) from adult Col1a1-NO66 transgenic mice showed an increase in adipogenesis and a decrease in osteogenesis. Taken together, these data demonstrate a crucial role for NO66 in adult bone formation and homeostasis. Our Col1a1-NO66 transgenic mice provide a novel animal model for the mechanistic and therapeutic study of NO66 in osteoporosis.

2.
Cell Death Dis ; 12(6): 521, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-34021120

RESUMEN

The developmental origins of mesenchymal progenitor cells (MPCs) and molecular machineries regulating their fate and differentiation are far from defined owing to their complexity. Osteoblasts and adipocytes are descended from common MPCs. Their fates are collectively determined by an orchestra of pathways in response to physiological and external cues. The canonical Wnt pathway signals MPCs to commit to osteogenic differentiation at the expense of adipogenic fate. In contrast to ß-catenin, p53's anti-osteogenic function is much less understood. Both activities are thought to be achieved through targeting Runx2 and/or Osterix (Osx, Sp7) transcription. Precisely, how Osx activity is dictated by ß-catenin or p53 is not clarified and represents a knowledge gap that, until now, has largely been taken for granted. Using conditional lineage-tracing mice, we demonstrated that chondrocytes gave rise to a sizable fraction of MPCs, which served as progenitors of chondrocyte-derived osteoblasts (Chon-ob). Wnt/ß-catenin activity was only required at the stage of chondrocyte-derived mesenchymal progenitor (C-MPC) to Chon-ob differentiation. ß-catenin- C-MPCs lost osteogenic ability and favored adipogenesis. Mechanistically, we discovered that p53 activity was elevated in ß-catenin- MPCs including ß-catenin- C-MPCs and deleting p53 from the ß-catenin- MPCs fully restored osteogenesis. While high levels of p53 were present in the nuclei of ß-catenin- MPCs, Osx was confined to the cytoplasm, implying a mechanism that did not involve direct p53-Osx interaction. Furthermore, we found that p53's anti-osteogenic activity was dependent on its DNA-binding ability. Our findings identify chondrocytes as an additional source for MPCs and indicate that Wnt/ß-catenin discretely regulates chondrocyte to C-MPC and the subsequent C-MPC to osteoblast developments. Most of all we unveil a previously unrecognized functional link between ß-catenin and p53, placing p53's negative role in the context of Wnt/ß-catenin signaling-induced MPC osteogenic differentiation.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Vía de Señalización Wnt/fisiología , Adipogénesis/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Condrogénesis/genética , Regulación hacia Abajo/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/fisiología , Osteogénesis/genética , Proteína p53 Supresora de Tumor/genética , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
3.
J Bone Miner Res ; 33(6): 1126-1140, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29405385

RESUMEN

The Sp7/Osterix transcription factor is essential for bone development. Mutations of the Sp7 gene in humans are associated with craniofacial anomalies and osteogenesis imperfecta. However, the role of Sp7 in embryonic tooth development remains unknown. Here we identified the functional requirement of Sp7 for dentin synthesis and tooth development. Sp7-null mice exhibit craniofacial dysmorphogenesis and are completely void of alveolar bone. Surprisingly, initial tooth morphogenesis progressed normally in Sp7-null mice. Thus the formation of alveolar bone is not a prerequisite for tooth morphogenesis. Sp7 is required for mineralization of palatal tissue but is not essential for palatal fusion. The reduced proliferative capacity of Sp7-deficient ectomesenchyme results in small and misshapen teeth with randomly arranged cuboidal preodontoblasts and preameloblasts. Sp7 promotes functional maturation and polarization of odontoblasts. Markers of mature odontoblast (Col1a, Oc, Dspp, Dmp1) and ameloblast (Enam, Amelx, Mmp20, Amtn, Klk4) are barely expressed in incisors and molar tissues of Sp7-null mice. Consequently, dentin and enamel matrix are absent in the Sp7-null littermates. Interestingly, the Sp7 expression is restricted to cells of the dental mesenchyme indicating the effect on oral epithelium-derived ameloblasts is cell-nonautonomous. Abundant expression of Fgf3 and Fgf8 ligand was noted in the developing tooth of wild-type mice. Both ligands were remarkably absent in the Sp7-null incisor and molar, suggesting cross-signaling between mesenchyme and epithelium is disrupted. Finally, promoter-reporter assays revealed that Sp7 directly controls the expression of Fgf-ligands. Together, our data demonstrate that Sp7 is obligatory for the differentiation of both ameloblasts and odontoblasts but not for the initial tooth morphogenesis. © 2018 American Society for Bone and Mineral Research.


Asunto(s)
Ameloblastos/citología , Ameloblastos/metabolismo , Diferenciación Celular , Odontoblastos/citología , Odontoblastos/metabolismo , Factor de Transcripción Sp7/metabolismo , Animales , Animales Recién Nacidos , Calcificación Fisiológica , Proliferación Celular , Colágeno/metabolismo , Dentina/metabolismo , Desarrollo Embrionario , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Incisivo/crecimiento & desarrollo , Incisivo/metabolismo , Incisivo/ultraestructura , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Morfogénesis , Hueso Paladar/metabolismo , Transducción de Señal , Factor de Transcripción Sp7/deficiencia , Factor de Transcripción Sp7/genética , Células Madre/metabolismo
4.
Methods Mol Biol ; 1627: 139-161, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28836200

RESUMEN

Excessive deposition of extracellular matrix (ECM) is a common hallmark of fibrotic diseases in various organs. Chiefly among this ECM are collagen types I and III, secreted by local fibroblasts, and other mesenchymal cells recruited for repair purposes. In the last two decades, the search for a fibroblast-specific promoter/enhancer has intensified in order to control the regulation of ECM in these cells and limit the scarring of the fibrotic process. In our previous work, we characterized an enhancer region 17 kb upstream of the Col1a2 gene transcription start site. This enhancer in transgenic mice is expressed mainly in mesenchymal cells during development and in adults upon injury. When driving transgenes such as beta-galactosidase or luciferase, this construct acts as an informative reporter of collagen transcription and is predictive of collagen type I deposition. In this chapter, we provide detailed protocols for identifying similar enhancers and using the sequence to generate a construct for transfection and producing transgenic animals. We also provided information on the use of luminescence in transgenic mice, tissue processing, as well as using cre/lox system to obtain conditional gain and loss of function in mice.


Asunto(s)
Colágeno Tipo I/genética , Elementos de Facilitación Genéticos , Expresión Génica , Genes Reporteros , Células Madre Mesenquimatosas/metabolismo , Regiones Promotoras Genéticas , Animales , Clonación Molecular , Biología Computacional/métodos , Bases de Datos de Ácidos Nucleicos , Fibroblastos/metabolismo , Mediciones Luminiscentes/métodos , Ratones , Ratones Transgénicos , Imagen Molecular , Ratas , Transfección , Transgenes , Navegador Web , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
5.
J Biol Chem ; 292(2): 528-538, 2017 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-27881681

RESUMEN

Type II collagen α1 is specific for cartilaginous tissues, and mutations in its gene are associated with skeletal diseases. Its expression has been shown to be dependent on SOX9, a master transcription factor required for chondrogenesis that binds to an enhancer region in intron 1. However, ChIP sequencing revealed that SOX9 does not strongly bind to intron 1, but rather it binds to intron 6 and a site 30 kb upstream of the transcription start site. Here, we aimed to determine the role of the novel SOX9-binding site in intron 6. We prepared reporter constructs that contain a Col2a1 promoter, intron 1 with or without intron 6, and the luciferase gene. Although the reporter constructs were not activated by SOX9 alone, the construct that contained both introns 1 and 6 was activated 5-10-fold by the SOX9/SOX5 or the SOX9/SOX6 combination in transient-transfection assays in 293T cells. This enhancement was also observed in rat chondrosarcoma cells that stably expressed the construct. CRISPR/Cas9-induced deletion of intron 6 in RCS cells revealed that a 10-bp region of intron 6 is necessary both for Col2a1 expression and SOX9 binding. Furthermore, SOX9, but not SOX5, binds to this region as demonstrated in an electrophoretic mobility shift assay, although both SOX9 and SOX5 bind to a larger 325-bp fragment of intron 6 containing this small sequence. These findings suggest a novel mechanism of action of SOX5/6; namely, the SOX9/5/6 combination enhances Col2a1 transcription through a novel enhancer in intron 6 together with the enhancer in intron 1.


Asunto(s)
Colágeno Tipo II/biosíntesis , Elementos de Facilitación Genéticos/fisiología , Regulación de la Expresión Génica/fisiología , Intrones/fisiología , Factor de Transcripción SOX9/metabolismo , Animales , Línea Celular Tumoral , Colágeno Tipo II/genética , Humanos , Ratones , Ratas , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Transcripción Genética/fisiología
6.
Sci Rep ; 6: 29916, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27436052

RESUMEN

Several lines of evidence indicate that connective tissue growth factor (CTGF/CCN2) stimulates chondrocyte proliferation and maturation. Given the fact that SOX9 is essential for several steps of the chondrocyte differentiation pathway, we asked whether Ctgf (Ccn2) is the direct target gene of SOX9. We found that Ctgf mRNA was down-regulated in primary sternal chondrocytes from Sox9(flox/flox) mice infected with Ad-CMV-Cre. We performed ChIP-on-chip assay using anti-SOX9 antibody, covering the Ctgf gene from 15 kb upstream of its 5'-end to 10 kb downstream of its 3'-end to determine SOX9 interaction site. One high-affinity interaction site was identified in the Ctgf proximal promoter by ChIP-on-chip assay. An important SOX9 regulatory element was found to be located in -70/-64 region of the Ctgf promoter. We found the same site for SOX9 binding to the Ctgf promoter in nucleus pulposus (NP) cells. The loss of Sox9 in growth plate chondrocytes in knee joint and in NP cells in intervertebral disc led to the decrease in CTGF expression. We suggest that Ctgf is the direct target gene of SOX9 in chondrocytes and NP cells. Our study establishes a strong link between two regulatory molecules that have a major role in cartilaginous tissues.


Asunto(s)
Condrocitos/citología , Condrocitos/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Placa de Crecimiento/citología , Núcleo Pulposo/citología , Factor de Transcripción SOX9/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Extremidades , Eliminación de Gen , Humanos , Ratones Noqueados , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Esternón/citología
8.
J Bone Miner Res ; 30(9): 1608-17, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25736226

RESUMEN

Our previous studies indicated that the Jumonji C (JmjC)-domain-containing NO66 is a histone demethylase with specificity for methylated histone H3K4 and H3K36. NO66 binds to the transcription factor Osterix (Osx) and inhibits its transcriptional activity in promoter assays. However, the physiological role of NO66 in formation of mammalian bones is unknown. Here, using a genetically engineered mouse model, we show that during early skeletal development, Prx1-Cre-dependent mesenchymal deletion of NO66 promotes osteogenesis and formation of both endochondral as well as intramembranous skeletal elements, leading to a larger skeleton and a high bone mass phenotype in adult mice. The excess bone formation in mice where NO66 was deleted in cells of mesenchymal origin is associated with an increase in the number of preosteoblasts and osteoblasts. Further analysis revealed that in the embryonic limbs and adult calvaria of mice with deletion of NO66 in cells of mesenchymal origin, expression of several genes including bone morphogenetic protein 2 (Bmp2), insulin-like growth factor 1 (Igf1), and osteoclast inhibitor osteoprotegerin was increased, concurrent with an increase in expression of bone formation markers such as osterix (Osx), type I collagen, and bone sialoprotein (Bsp). Taken together, our results provide the first in vivo evidence that NO66 histone demethylase plays an important role in mammalian osteogenesis during early development as well as in adult bone homeostasis. We postulate that NO66 regulates bone formation, at least in part, via regulating the number of bone-forming cells and expression of multiple genes that are critical for these processes.


Asunto(s)
Huesos/metabolismo , Eliminación de Gen , Histona Demetilasas con Dominio de Jumonji/metabolismo , Mesodermo/metabolismo , Osteogénesis/genética , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular , Colágeno Tipo I/metabolismo , Femenino , Fluoresceínas/química , Regulación de la Expresión Génica , Genotipo , Histonas/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Sialoproteína de Unión a Integrina/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Masculino , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Fenotipo , Factor de Transcripción Sp7 , Microtomografía por Rayos X
9.
FASEB J ; 29(6): 2555-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25746793

RESUMEN

Previous studies showed that nucleolar protein 66 (NO66), the Jumonji C-domain-containing histone demethylase for methylated histone H3K4 and H3K36 (H3K36me), negatively regulates osteoblast differentiation in vitro by inhibiting the activity of transcription factor osterix (Osx). However, whether NO66 affects mammalian skeletogenesis in vivo is not yet known. Here, we generated transgenic (TG) mice overexpressing a flag-tagged NO66 transgene driven by the Prx1 (paired related homeobox 1) promoter. We found that NO66 overexpression in Prx1-expressing mesenchymal cells inhibited skeletal growth and bone formation. The inhibitory phenotype was associated with >50% decreases in chondrocyte/osteoblast proliferation and differentiation. Moreover, we found that in bones of NO66-TG mice, expression of Igf1, Igf1 receptor (Igf1r), runt-related transcription factor 2, and Osx was significantly down-regulated (P < 0.05). Consistent with these results, we observed >50% reduction in levels of phosphorylated protein kinase B (Akt) and H3K36me3 in bones of NO66-TG mice, suggesting an inverse correlation between NO66 histone demethylase and the activity of IGF1R/Akt signaling. This correlation was further confirmed by in vitro assays of C2C12 cells with NO66 overexpression. We propose that the decrease in the IGF1R/Akt signaling pathway in mice with mesenchymal overexpression of NO66 may contribute in part to the inhibition of skeletal growth and bone formation.


Asunto(s)
Huesos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Mesodermo/metabolismo , Cráneo/metabolismo , Animales , Western Blotting , Huesos/citología , Huesos/embriología , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/genética , Condrocitos/citología , Condrocitos/metabolismo , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Mesodermo/citología , Mesodermo/embriología , Ratones Transgénicos , Microscopía Fluorescente , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Cráneo/embriología , Factor de Transcripción Sp7 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
PLoS Genet ; 10(12): e1004820, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25474590

RESUMEN

One of the crucial steps in endochondral bone formation is the replacement of a cartilage matrix produced by chondrocytes with bone trabeculae made by osteoblasts. However, the precise sources of osteoblasts responsible for trabecular bone formation have not been fully defined. To investigate whether cells derived from hypertrophic chondrocytes contribute to the osteoblast pool in trabecular bones, we genetically labeled either hypertrophic chondrocytes by Col10a1-Cre or chondrocytes by tamoxifen-induced Agc1-CreERT2 using EGFP, LacZ or Tomato expression. Both Cre drivers were specifically active in chondrocytic cells and not in perichondrium, in periosteum or in any of the osteoblast lineage cells. These in vivo experiments allowed us to follow the fate of cells labeled in Col10a1-Cre or Agc1-CreERT2 -expressing chondrocytes. After the labeling of chondrocytes, both during prenatal development and after birth, abundant labeled non-chondrocytic cells were present in the primary spongiosa. These cells were distributed throughout trabeculae surfaces and later were present in the endosteum, and embedded within the bone matrix. Co-expression studies using osteoblast markers indicated that a proportion of the non-chondrocytic cells derived from chondrocytes labeled by Col10a1-Cre or by Agc1-CreERT2 were functional osteoblasts. Hence, our results show that both chondrocytes prior to initial ossification and growth plate chondrocytes before or after birth have the capacity to undergo transdifferentiation to become osteoblasts. The osteoblasts derived from Col10a1-expressing hypertrophic chondrocytes represent about sixty percent of all mature osteoblasts in endochondral bones of one month old mice. A similar process of chondrocyte to osteoblast transdifferentiation was involved during bone fracture healing in adult mice. Thus, in addition to cells in the periosteum chondrocytes represent a major source of osteoblasts contributing to endochondral bone formation in vivo.


Asunto(s)
Transdiferenciación Celular , Condrocitos/fisiología , Curación de Fractura/fisiología , Crecimiento y Desarrollo , Osteoblastos/fisiología , Animales , Desarrollo Óseo/fisiología , Cartílago/crecimiento & desarrollo , Transdiferenciación Celular/genética , Células Cultivadas , Condrogénesis/fisiología , Embrión de Mamíferos , Desarrollo Embrionario/genética , Femenino , Placa de Crecimiento/embriología , Placa de Crecimiento/metabolismo , Crecimiento y Desarrollo/genética , Ratones , Ratones Transgénicos , Osteogénesis/fisiología , Embarazo
11.
Cell Rep ; 9(2): 581-90, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25310984

RESUMEN

Hematopoietic stem cells (HSCs) reside within a specialized niche where interactions with vasculature, osteoblasts, and stromal components regulate their self-renewal and differentiation. Little is known about bone marrow niche formation or the role of its cellular components in HSC development; therefore, we established the timing of murine fetal long bone vascularization and ossification relative to the onset of HSC activity. Adult-repopulating HSCs emerged at embryonic day 16.5 (E16.5), coincident with marrow vascularization, and were contained within the c-Kit(+)Sca-1(+)Lin(-) (KSL) population. We used Osterix-null (Osx(-/-)) mice that form vascularized marrow but lack osteolineage cells to dissect the role(s) of these cellular components in HSC development. Osx(-/-) fetal bone marrow cells formed multilineage colonies in vitro but were hyperproliferative and failed to home to and/or engraft transplant recipients. Thus, in developing bone marrow, the vasculature can sustain multilineage progenitors, but interactions with osteolineage cells are needed to regulate long-term HSC proliferation and potential.


Asunto(s)
Médula Ósea/embriología , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Osteogénesis , Nicho de Células Madre , Animales , Médula Ósea/irrigación sanguínea , Linaje de la Célula , Proliferación Celular , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Transcripción Sp7 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
PLoS One ; 9(9): e107577, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25229425

RESUMEN

The transcription factor SOX9 plays an essential role in determining the fate of several cell types and is a master factor in regulation of chondrocyte development. Our aim was to determine which genes in the genome of chondrocytes are either directly or indirectly controlled by SOX9. We used RNA-Seq to identify genes whose expression levels were affected by SOX9 and used SOX9 ChIP-Seq to identify those genes that harbor SOX9-interaction sites. For RNA-Seq, the RNA expression profile of primary Sox9flox/flox mouse chondrocytes infected with Ad-CMV-Cre was compared with that of the same cells infected with a control adenovirus. Analysis of RNA-Seq data indicated that, when the levels of Sox9 mRNA were decreased more than 8-fold by infection with Ad-CMV-Cre, 196 genes showed a decrease in expression of at least 4-fold. These included many cartilage extracellular matrix (ECM) genes and a number of genes for ECM modification enzymes (transferases), membrane receptors, transporters, and others. In ChIP-Seq, 75% of the SOX9-interaction sites had a canonical inverted repeat motif within 100 bp of the top of the peak. SOX9-interaction sites were found in 55% of the genes whose expression was decreased more than 8-fold in SOX9-depleted cells and in somewhat fewer of the genes whose expression was reduced more than 4-fold, suggesting that these are direct targets of SOX9. The combination of RNA-Seq and ChIP-Seq has provided a fuller understanding of the SOX9-controlled genetic program of chondrocytes.


Asunto(s)
Condrocitos/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Factor de Transcripción SOX9/metabolismo , Animales , Sitios de Unión , Inmunoprecipitación de Cromatina , Expresión Génica , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Ratones Noqueados , Motivos de Nucleótidos , Posición Específica de Matrices de Puntuación , Unión Proteica , Transporte de Proteínas , Factor de Transcripción SOX9/genética
13.
Connect Tissue Res ; 55 Suppl 1: 83-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25158187

RESUMEN

Runx2 and Sp7 transcription factors are essential for skeletogenesis. Targeted deletion of either gene results in failure of osteoblast differentiation and bone formation. Loss of bone-matrix gene expression is surprisingly similar in Sp7 and Runx2 null mice. The molecular mechanisms responsible for similar transcriptional regulation of target genes remain largely unknown. Here, we demonstrate that Runx2 and Sp7 interact physically and functionally. Both proteins are co-expressed in osteoblastic cells. We first characterized a panel of Sp7 antibodies and demonstrate that majority of the published antibodies do not recognize Sp7 protein. Co-immunoprecipitation studies revealed that endogenous Runx2 protein physically interacts with Sp7 protein. We identified that runt homology domain (RHD) of Runx2 protein is involved in physical association with Sp7. Functional consequences of Runx2-Sp7 physical interaction was then assessed by promoter-reporter assays. We selected promoters of osteocalcin (OC), a marker of mature osteoblast and fibroblast growth factor 3 (FGF3), a signaling molecule that determine the fate of embryonic ecto-mesenchyme. Runx2 and Sp7 stimulate OC-promoter activity by 3-folds in epithelial cells. However, when both proteins were co-expressed, a dose-dependent synergistic activation of 22-folds was noted. Similar pattern of synergistic activation of OC-promoter was noted in mesenchymal cell. FGF3 promoter was activated by 25 - and 30-folds with Runx2 and Sp7 respectively. Again a dose-dependent synergistic activation of 130-folds was evident when Runx2 and Sp7 were co-expressed in epithelial cells. Synergistic activation of FGF3 promoter was also noted in mesenchymal cells. Together, our data demonstrated that Runx2-Sp7 molecular complex functionally cooperate for maximal induction of cell-phenotype-restricted genes.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Osteoblastos/citología , Osteocalcina/metabolismo , Osteogénesis/fisiología , Factores de Transcripción/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Regulación de la Expresión Génica/fisiología , Humanos , Unión Proteica , Factor de Transcripción Sp7
14.
J Biol Chem ; 288(49): 35138-48, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24155239

RESUMEN

SOX9 is a transcription factor that acts as a key regulator at various stages of cartilage differentiation. There is ample evidence that intracellular SOX9 protein levels are tightly regulated both by sumoylation and by degradation through the ubiquitin-proteasome pathway. Using a proteomics approach, here we report the identification of a SOX9-binding protein, E6-AP/UBE3A, that may act as a ubiquitin ligase toward Sox9. E6-AP bound SOX9 through the region consisting mostly of its high mobility group domain in vitro. In nuclear lysates, FLAG-tagged E6-AP coprecipitated with Sox9 and its high mobility group domain. This finding was estimated using nuclear lysates from a chondrocytic cell line that endogenously expresses E6-AP and SOX9. Accordingly, ectopically expressed E6-AP and SOX9 colocalized in the nucleus. We show that E6-AP ubiquitinates SOX9 in vitro and in vivo and that SOX9 levels are enhanced after addition of the proteasome inhibitor bortezomib. Similar, siRNA knockdown of E6-AP and the E2 ligase Ubc9 increased cellular SOX9 amounts, supporting the notion that SOX9 may be ubiquitinated in hypertrophic chondrocytes by E6-AP and degraded by proteasomes. This is in accordance with the distribution of SOX9 levels, which are high in proliferating and prehypertrophic chondrocytes but low in hypertrophic chondrocytes, whereas E6-AP levels are high in hypertrophic chondrocytes and low in prehypertrophic chondrocytes. Furthermore, E6-AP-deficient mice showed SOX9 accumulation in chondrocytes and the brain. These findings support the concept that E6-AP regulates SOX9 levels in developing cartilage by acting as a ubiquitin ligase.


Asunto(s)
Factor de Transcripción SOX9/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Bovinos , Línea Celular , Chlorocebus aethiops , Condrocitos/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Unión Proteica , Proteómica , ARN Interferente Pequeño/genética , Factor de Transcripción SOX9/química , Factor de Transcripción SOX9/genética , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
15.
PLoS One ; 8(7): e69859, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23922826

RESUMEN

Osterix (Osx) is an essential transcription factor for osteoblast differentiation and bone formation. Osx knockout show a complete absence of bone formation, whereas Osx conditional knockout in osteoblasts produce an osteopenic phenotype after birth. Here, we questioned whether Osx has a potential role in regulating physiological homeostasis. In Osx heterozygotes expressing low levels of Osx in bones, the expression levels of pro-inflammatory cytokines were significantly elevated, indicating that reduced Osx expression may reflect an inflammatory-prone state. In particular, the expression of interleukin-6, a key mediator of chronic inflammation, was increased in Osx heterozygotes and decreased in Osx overexpressing osteoblasts, and transcriptionally down-regulated by Osx. Although no significant differences were revealed in renal morphology and function between Osx heterozygotes and wild-type under normoxic conditions, recovery of kidneys after ischemic damage was remarkably delayed in Osx heterozygotes, as indicated by elevated blood urea nitrogen and creatinine levels, and by morphological alterations consistent with acute tubular necrosis. Eventually, protracted low Osx expression level caused an inflammatory-prone state in the body, resulting in the enhanced susceptibility to renal injury and the delayed renal repair after ischemia/reperfusion. This study suggests that the maintenance of Osx expression in bone is important in terms of preventing the onset of an inflammatory-prone state.


Asunto(s)
Interleucina-6/biosíntesis , Riñón/metabolismo , Riñón/patología , Regeneración , Factores de Transcripción/metabolismo , Animales , Línea Celular , Regulación hacia Abajo , Heterocigoto , Inflamación/patología , Interleucina-6/genética , Riñón/fisiopatología , Pruebas de Función Renal , Ratones , Modelos Biológicos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteogénesis , Factor de Transcripción Sp7 , Transcripción Genética
16.
J Biol Chem ; 288(23): 16430-16437, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23620590

RESUMEN

Osterix (Osx) is an osteoblast-specific transcriptional factor and is required for osteoblast differentiation and bone formation. A JmjC domain-containing protein NO66 was previously found to participate in regulation of Osx transcriptional activity and plays an important role in osteoblast differentiation through interaction with Osx. Here, we report the crystal structure of NO66 forming in a functional tetramer. A hinge domain links the N-terminal JmjC domain and C-terminal winged helix-turn-helix domain of NO66, and both domains are essential for tetrameric assembly. The oligomerization interface of NO66 interacts with a conserved fragment of Osx. We show that the hinge domain-dependent oligomerization of NO66 is essential for inhibition of Osx-dependent gene activation. Our findings suggest that homo-oligomerization of JmjC domain containing proteins might play a physiological role through interactions with other regulatory factors during gene expression.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Cromosómicas no Histona , Regulación de la Expresión Génica/fisiología , Osteoblastos/metabolismo , Multimerización de Proteína/fisiología , Proteínas Represoras , Factores de Transcripción , Línea Celular , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Cristalografía por Rayos X , Dioxigenasas , Secuencias Hélice-Giro-Hélice , Histona Demetilasas , Humanos , Osteoblastos/citología , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción Sp7 , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología
17.
PLoS One ; 8(3): e58104, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23472141

RESUMEN

Osterix (Osx) is an osteoblast-specific transcription factor which is essential for bone formation. MicroRNAs (miRNAs) have been previously shown to be involved in osteogenesis. However, it is unclear whether Osx is involved in the regulation of miRNA expression. In this study, we have identified groups of miRNAs that are differentially expressed in calvaria of the E18.5 Osx(-/-) embryos compared to wild type embryos. The correlation between the levels of miRNAs and Osx expression was further verified in cultured M-Osx cells in which over-expression of Osx is inducible. Our results suggest that Osx down-regulates expression of a group of miRNAs including mir-133a and -204/211, but up-regulates expression of another group of miRNAs such as mir-141/200a. Mir-133a and -204/211 are known to target the master osteogenic transcription factor Runx2. Further assays suggest that Sost, which encodes the Wnt signaling antagonist Sclerostin, and alkaline phosphatase (ALP) are two additional targets of mir-204/211. Mir-141/200a has been known to target the transcription factor Dlx5. Thus, we postulate that during the process of Osx-controlled osteogenesis, Osx has the ability to coordinately modulate Runx2, Sclerostin, ALP and Dlx5 proteins at levels appropriate for optimal osteoblast differentiation and function, at least in part, through regulation of specific miRNAs. Our study shows a tight correlation between Osx and the miRNAs involved in bone formation, and provides new information about molecular mechanisms of Osx-controlled osteogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Osteogénesis/genética , Cráneo/metabolismo , Factores de Transcripción/fisiología , Proteínas Adaptadoras Transductoras de Señales , Fosfatasa Alcalina/metabolismo , Animales , Genotipo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Noqueados , Transducción de Señal , Factor de Transcripción Sp7 , Factores de Tiempo , Factores de Transcripción/genética
18.
J Cell Sci ; 126(Pt 10): 2164-75, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23525012

RESUMEN

Connective tissue growth factor (CTGF) plays an important role in the pathogenesis of chronic fibrotic diseases. However, the mechanism by which paracrine effects of CTGF control the cell fate of neighboring epithelial cells is not known. In this study, we investigated the paracrine effects of CTGF overexpressed in fibroblasts of Col1a2-CTGF transgenic mice on epithelial cells of skin and lung. The skin and lungs of Col1a2-CTGF transgenic mice were examined for phenotypic markers of epithelial activation and differentiation and stimulation of signal transduction pathways. In addition to an expansion of the dermal compartment in Col1a2-CTGF transgenic mice, the epidermis was characterized by focal hyperplasia, and basal cells stained positive for αSMA, Snail, S100A4 and Sox9, indicating that these cells had undergone a change in their genetic program. Activation of phosphorylated p38 and phosphorylated Erk1/2 was observed in the granular and cornified layers of the skin. Lung fibrosis was associated with a marked increase in cells co-expressing epithelial and mesenchymal markers in the lesional and unaffected lung tissue of Col1a2-CTGF mice. In epithelial cells treated with TGFß, CTGF-specific siRNA-mediated knockdown suppressed Snail, Sox9, S100A4 protein levels and restored E-cadherin levels. Both adenoviral expression of CTGF in epithelial cells and treatment with recombinant CTGF induced EMT-like morphological changes and expression of α-SMA. Our in vivo and in vitro data supports the notion that CTGF expression in mesenchymal cells in the skin and lungs can cause changes in the differentiation program of adjacent epithelial cells. We speculate that these changes might contribute to fibrogenesis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Transición Epitelial-Mesenquimal , Fibroblastos/fisiología , Hiperplasia Epitelial Focal/fisiopatología , Fibrosis Pulmonar/fisiopatología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Colágeno Tipo I/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Transición Epitelial-Mesenquimal/genética , Pulmón/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Transgénicos , Comunicación Paracrina , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Piel/patología , Factor de Crecimiento Transformador beta/inmunología , Transgenes/genética
19.
Biochem Biophys Res Commun ; 432(1): 188-92, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23313488

RESUMEN

Osx plays essential roles in regulating osteoblast and chondrocyte differentiation, and bone formation during mouse skeletal development. However, many questions remain regarding the requirement for Osx in different cell lineages. In this study, we asked whether Osx is required for craniofacial bone formation derived from cranial neural crest (CNC) cells. The Osx gene was conditionally inactivated in CNC-derived cells using a Wnt1-Cre recombination system. Neural crest-specific inactivation of Osx resulted in the complete absence of intramembranous skeletal elements derived from the CNC, and CNC-derived endochondral skeletal elements were also affected by Osx inactivation. Interestingly, Osx inactivated CNC-derived cells, which were recapitulated by lacZ expression, occupied the same regions of craniofacial skeletal elements as observed for controls. However, cells lost their osteogenic ability to differentiate into functional osteoblasts by Osx inactivation. These results suggest that Osx is important for craniofacial bone formation by CNC-derived cells. This finding provides novel insights of the regulation of craniofacial development by the gene network and transcription factors, and the understanding of human diseases caused by neural crest developmental abnormalities.


Asunto(s)
Anomalías Craneofaciales/genética , Huesos Faciales/embriología , Cresta Neural/anomalías , Osteogénesis/genética , Factores de Transcripción/fisiología , Animales , Anomalías Craneofaciales/patología , Huesos Faciales/anomalías , Huesos Faciales/patología , Silenciador del Gen , Integrasas/genética , Ratones , Ratones Transgénicos , Factor de Transcripción Sp7 , Factores de Transcripción/genética , Proteína Wnt1/genética
20.
J Bone Miner Res ; 27(12): 2511-25, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22777888

RESUMEN

Sox9 is an essential transcription factor for the differentiation of the chondrocytic lineage during embryonic development. To test whether Sox9 continues to play a critical role in cartilaginous tissues in the adult mice, we used an inducible, genetic strategy to disrupt the Sox9 gene postnatally in these tissues. The postnatal inactivation of Sox9 led to stunted growth characterized by decreased proliferation, increased cell death, and dedifferentiation of growth plate chondrocytes. Upon postnatal Sox9 inactivation in the articular cartilage, the sulfated proteoglycan and aggrecan content of the uncalcified cartilage were rapidly depleted and the degradation of aggrecan was accompanied by higher ADAMTS5 immunostaining and increased detection of the aggrecan neoepitope, NITEGE. In spite of the severe loss of Collagen 2a1 mRNA, the Collagen II protein persisted in the articular cartilage, and no histopathological signs of osteoarthritis were observed. The homeostasis of the intervertebral disk (IVD) was dramatically altered upon Sox9 depletion, resulting in disk compression and subsequent degeneration. Inactivation of Sox9 in the IVD markedly reduced the expression of several genes encoding extracellular matrix proteins, as well as some of the enzymes responsible for their posttranslational modification. Furthermore, the loss of Sox9 in the IVD decreased the expression of cytokines, cell-surface receptors, and ion channels, suggesting that Sox9 coordinates a large genetic program that is instrumental for the proper homeostasis of the cells contained in the IVD postnatally. Our results indicate that Sox9 has an essential role in the physiological control of cartilaginous tissues in adult mice. © 2012 American Society for Bone and Mineral Research.


Asunto(s)
Cartílago Articular/metabolismo , Factor de Transcripción SOX9/fisiología , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Colágeno Tipo II/metabolismo , Placa de Crecimiento/metabolismo , Disco Intervertebral/efectos de los fármacos , Masculino , Ratones , Fenotipo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...