Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Dev Dyn ; 248(10): 1009-1019, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31397024

RESUMEN

BACKGROUND: The murine calvaria has several membrane bones with different tissue origins (e.g., neural crest-derived frontal bone vs. mesoderm-derived parietal bone). Neural crest-derived frontal bone exhibits superior osteogenic activities and bone regeneration. MicroRNA (miRNA) has been emerged as a crucial regulator during organogenesis and is involved in a range of developmental processes. However, the underlying roles of miRNA regulation in frontal bone and parietal bone is unknown. RESULTS: Total of 83 significantly expressed known miRNAs were identified in frontal bones versus parietal bones. The significantly enriched gene ontology and KEGG pathway that were predicted by the enrichment miRNAs were involved in several biological processes (cell differentiation, cell adhesion, and transcription), and multiple osteogenic pathways (e.g., focal adhesion, MAPK, VEGF, Wnt, and insulin signaling pathway. Focal adhesion and insulin signaling pathway were selected for target verification and functional analysis, and several genes were predicted to be targets genes by the differentially expressed miRNAs, and these targets genes were tested with significant expressions. CONCLUSIONS: Our results revealed a novel pattern of miRNAs in murine calvaria with dual tissue origins, and explorations of these miRNAs will be valuable for the translational studies to enhance osteogenic potential and bone regeneration in the clinic.


Asunto(s)
Hueso Frontal/metabolismo , MicroARNs/análisis , Hueso Parietal/metabolismo , Cráneo/metabolismo , Animales , Regeneración Ósea , Adhesiones Focales , Insulina/metabolismo , Ratones , MicroARNs/fisiología , Osteogénesis , Transducción de Señal
2.
Med Mol Morphol ; 52(3): 156-163, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30499042

RESUMEN

Most facial bones, including frontal bones, are derived from neural crest cells through intramembranous ossification. Fibroblast growth factor receptor 1 (Fgfr1) plays a pivotal role in craniofacial bone development, and loss of Fgfr1 leads to cleft palate and facial cleft defects in newborn mice. However, the potential role of the Fgfr1 gene in neural crest cell-mediated craniofacial development remains unclear. To investigate the role of Fgfr1 in neural crest cells, we analyzed Wnt1-Cre;Fgfr1flox/flox mice. Our results show that specific knockout of Fgfr1 in neural crest cells induced heterotopic chondrogenesis and osteogenesis at the interface of the anterior portions of frontal bones. We observed that heterotopic bone formation continued through postnatal day 28, whereas heterotopic chondrogenesis lasted only through the embryonic period. In summary, our results indicate that loss of Fgfr1 in neural crest cells leads to heterotopic chondrogenesis and osteogenesis.


Asunto(s)
Condrogénesis , Hueso Frontal/crecimiento & desarrollo , Cresta Neural/crecimiento & desarrollo , Osteogénesis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Hueso Frontal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Cresta Neural/citología , Cresta Neural/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
3.
BMC Dev Biol ; 16(1): 37, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27756203

RESUMEN

BACKGROUND: Increased apposition of the frontal and parietal bones of the skull during embryogenesis may be a risk factor for the subsequent development of premature skull fusion, or craniosynostosis. Human craniosynostosis is a prevalent, and often serious embryological and neonatal pathology. Other than known mutations in a small number of contributing genes, the aetiology of craniosynostosis is largely unknown. Therefore, the identification of novel genes which contribute to normal skull patterning, morphology and premature suture apposition is imperative, in order to fully understand the genetic regulation of cranial development. RESULTS: Using advanced imaging techniques and quantitative measurement, we show that genetic deletion of the highly-conserved transcription factor Grainyhead-like 3 (Grhl3) in mice (Grhl3 -/- ) leads to decreased skull size, aberrant skull morphology and premature apposition of the coronal sutures during embryogenesis. Furthermore, Grhl3 -/- mice also present with premature collagen deposition and osteoblast alignment at the sutures, and the physical interaction between the developing skull, and outermost covering of the brain (the dura mater), as well as the overlying dermis and subcutaneous tissue, appears compromised in embryos lacking Grhl3. Although Grhl3 -/- mice die at birth, we investigated skull morphology and size in adult animals lacking one Grhl3 allele (heterozygous; Grhl3 +/- ), which are viable and fertile. We found that these adult mice also present with a smaller cranial cavity, suggestive of post-natal haploinsufficiency in the context of cranial development. CONCLUSIONS: Our findings show that our Grhl3 mice present with increased apposition of the frontal and parietal bones, suggesting that Grhl3 may be involved in the developmental pathogenesis of craniosynostosis.


Asunto(s)
Craneosinostosis/genética , Proteínas de Unión al ADN/genética , Hueso Frontal/metabolismo , Hueso Parietal/metabolismo , Factores de Transcripción/genética , Animales , Suturas Craneales/anomalías , Suturas Craneales/metabolismo , Craneosinostosis/embriología , Craneosinostosis/metabolismo , Proteínas de Unión al ADN/deficiencia , Desarrollo Embrionario/genética , Hueso Frontal/anomalías , Hueso Frontal/diagnóstico por imagen , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ratones Noqueados , Hueso Parietal/anomalías , Hueso Parietal/diagnóstico por imagen , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Factores de Riesgo , Cráneo/anomalías , Cráneo/metabolismo , Factores de Transcripción/deficiencia , Microtomografía por Rayos X
4.
Development ; 140(5): 1034-44, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23344708

RESUMEN

The mammalian skull vault consists of several intricately patterned bones that grow in close coordination. The growth of these bones depends on the precise regulation of the migration and differentiation of osteogenic cells from undifferentiated precursor cells located above the eye. Here, we demonstrate a role for Foxc1 in modulating the influence of Bmp signaling on the expression of Msx2 and the specification of these cells. Inactivation of Foxc1 results in a dramatic reduction in skull vault growth and causes an expansion of Msx2 expression and Bmp signaling into the area occupied by undifferentiated precursor cells. Foxc1 interacts directly with a Bmp responsive element in an enhancer upstream of Msx2, and acts to reduce the occupancy of P-Smad1/5/8. We propose that Foxc1 sets a threshold for the Bmp-dependent activation of Msx2, thus controlling the differentiation of osteogenic precursor cells and the rate and pattern of calvarial bone development.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Factores de Transcripción Forkhead/fisiología , Hueso Frontal/embriología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Animales , Desarrollo Óseo/genética , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/genética , Células Cultivadas , Embrión de Mamíferos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Hueso Frontal/crecimiento & desarrollo , Hueso Frontal/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteogénesis/genética , Osteogénesis/fisiología , Cráneo/embriología , Cráneo/metabolismo
5.
Stem Cells Dev ; 32(13-14): 398-409, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37078151

RESUMEN

The five flat bones of developing cranial plates are bounded by fibrous sutures, which remain open during development to accommodate for the growing brain. Kdm6A is a demethylase that removes the epigenetic repressive mark, trimethylated lysine 27 on histone 3 (H3K27me3), from the promoters of osteogenic genes, and has previously been reported to promote osteogenesis in cranial bone cells. This study generated a mesenchyme-specific deletion of a histone demethylase, Kdm6a, to assess the effects of Kdm6a loss, in cranial plate development and suture fusion. The results showed that the loss of Kdm6a in Prx1+ cranial cells caused increased anterior width and length in the calvaria of both male and female mice. However, the posterior length was further decreased in female mice. Moreover, loss of Kdm6a resulted in suppression of late suture development and calvarial frontal bone formation predominantly in female mice. In vitro assessment of calvaria cultures isolated from female Kdm6a knockout mice found significantly suppressed calvarial osteogenic differentiation potential, associated with decreased gene expression levels of Runx2 and Alkaline Phosphatase and increased levels of the suppressive mark, H3K27me3, on the respective gene promoters. Conversely, cultured calvaria bone cultures isolated from male Kdm6a knockout mice exhibited an increased osteogenic differentiation potential. Interestingly, the milder effects on cranial suture development in Kdm6a knockout male mice, were associated with an overcompensation of the Kdm6a Y-homolog, Kdm6c, and increased expression levels of Kdm6b in calvarial bone cultures. Taken together, these data demonstrate a role for Kdm6a during calvarial development and patterning, predominantly in female mice, and highlight the potential role of Kdm6 family members in patients with unexplained craniofacial deformities.


Asunto(s)
Suturas Craneales , Hueso Frontal , Animales , Femenino , Masculino , Ratones , Suturas Craneales/metabolismo , Hueso Frontal/metabolismo , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Ratones Noqueados , Osteogénesis/genética , Factores Sexuales
6.
Stem Cell Res Ther ; 13(1): 341, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35883153

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) leads to cell and tissue impairment, as well as functional deficits. Stem cells promote structural and functional recovery and thus are considered as a promising therapy for various nerve injuries. Here, we aimed to investigate the role of ectoderm-derived frontal bone mesenchymal stem cells (FbMSCs) in promoting cerebral repair and functional recovery in a murine TBI model. METHODS: A murine TBI model was established by injuring C57BL/6 N mice with moderate-controlled cortical impact to evaluate the extent of brain damage and behavioral deficits. Ectoderm-derived FbMSCs were isolated from the frontal bone and their characteristics were assessed using multiple differentiation assays, flow cytometry and microarray analysis. Brain repairment and functional recovery were analyzed at different days post-injury with or without FbMSC application. Behavioral tests were performed to assess learning and memory improvements. RNA sequencing analysis, immunofluorescence staining, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were used to examine inflammation reaction and neural regeneration. In vitro co-culture analysis and quantification of glutamate transportation were carried out to explore the possible mechanism of neurogenesis and functional recovery promoted by FbMSCs. RESULTS: Ectoderm-derived FbMSCs showed fibroblast like morphology and osteogenic differentiation capacity. FbMSCs were CD105, CD29 positive and CD45, CD31 negative. Different from mesoderm-derived MSCs, FbMSCs expressed the ectoderm-specific transcription factor Tfap2ß. TBI mice showed impaired learning and memory deficits. Microglia and astrocyte activation, as well as neural damage, were significantly increased post-injury. FbMSC application ameliorated the behavioral deficits of TBI mice and promoted neural regeneration. RNA sequencing analysis showed that signal pathways related to inflammation decreased, whereas those related to neural activation increased. Immunofluorescence staining and qRT-PCR data revealed that microglial activation and astrocyte polarization to the A1 phenotype were suppressed by FbMSC application. In addition, FGF1 secreted from FbMSCs enhanced glutamate transportation by astrocytes and alleviated the cytotoxic effect of excessive glutamate on neurons. CONCLUSIONS: Ectoderm-derived FbMSC application significantly alleviated neuroinflammation, brain injury, and excitatory toxicity to neurons, improved cognition and behavioral deficits in TBI mice. Therefore, ectoderm-derived FbMSCs could be ideal therapeutic candidates for TBI which mostly affect cells from the same embryonic origins as FbMSCs.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Células Madre Mesenquimatosas , Animales , Lesiones Encefálicas/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Ectodermo/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 1 de Crecimiento de Fibroblastos/uso terapéutico , Hueso Frontal/metabolismo , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Ácido Glutámico/uso terapéutico , Inflamación/metabolismo , Inflamación/terapia , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Enfermedades Neuroinflamatorias , Osteogénesis
7.
Genesis ; 48(11): 645-55, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20824629

RESUMEN

The Msx and Dlx families of homeobox proteins are important regulators for embryogenesis. Loss of Msx1 in mice results in multiple developmental defects including craniofacial malformations. Although Dlx5 is widely expressed during embryonic development, targeted null mutation of Dlx5 mainly affects the development of craniofacial bones. Msx1 and Dlx5 show overlapping expression patterns during frontal bone development. To investigate the functional significance of Msx1/Dlx5 interaction in regulating frontal bone development, we generated Msx1 and Dlx5 double null mutant mice. In Msx1(-/-) ;Dlx5(-/-) mice, the frontal bones defect was more severe than that of either Msx1(-/-) or Dlx5(-/-) mice. This aggravated frontal bone defect suggests that Msx1 and Dlx5 function synergistically to regulate osteogenesis. This synergistic effect of Msx1 and Dlx5 on the frontal bone represents a tissue specific mode of interaction of the Msx and Dlx genes. Furthermore, Dlx5 requires Msx1 for its expression in the context of frontal bone development. Our study shows that Msx1/Dlx5 interaction is crucial for osteogenic induction during frontal bone development.


Asunto(s)
Hueso Frontal/embriología , Proteínas de Homeodominio/fisiología , Factor de Transcripción MSX1/fisiología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos , Epistasis Genética/fisiología , Hueso Frontal/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Noqueados , Morfogénesis/genética , Morfogénesis/fisiología , Osteogénesis/genética , Osteogénesis/fisiología
8.
Dev Biol ; 331(2): 176-88, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414008

RESUMEN

Using a Cre-mediated conditional deletion approach, we have dissected the function of Twist1 in the morphogenesis of the craniofacial skeleton. Loss of Twist1 in neural crest cells and their derivatives impairs skeletogenic differentiation and leads to the loss of bones of the snout, upper face and skull vault. While no anatomically recognizable maxilla is formed, a malformed mandible is present. Since Twist1 is expressed in the tissues of the maxillary eminence and the mandibular arch, this finding suggests that the requirement for Twist1 is not the same in all neural crest derivatives. The effect of the loss of Twist1 function is not restricted to neural crest-derived bones, since the predominantly mesoderm-derived parietal and interparietal bones are also affected, presumably as a consequence of lost interactions with neural crest-derived tissues. In contrast, the formation of other mesodermal skeletal derivatives such as the occipital bones and most of the chondrocranium are not affected by the loss of Twist1 in the neural crest cells.


Asunto(s)
Morfogénesis/fisiología , Cresta Neural/embriología , Proteínas Nucleares/fisiología , Cráneo/embriología , Proteína 1 Relacionada con Twist/fisiología , Animales , Región Branquial/citología , Región Branquial/embriología , Región Branquial/fisiología , Hueso Frontal/embriología , Hueso Frontal/metabolismo , Maxilares/embriología , Maxilares/metabolismo , Ratones , Ratones Mutantes , Hueso Nasal/embriología , Hueso Nasal/metabolismo , Cresta Neural/citología , Cresta Neural/fisiología , Cráneo/citología , Cráneo/fisiología
9.
Science ; 175(4021): 544-6, 1972 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-5008597

RESUMEN

An enzymatic activity (procollagen peptidase), capable of converting the biosynthetic precursor procollagen to collagen at neutral pH, has been identified in rat and chick calvarial bone. Limited proteolysis of procollagen with chymotrypsin resulted in a similar transformation. The activity in bone can be demonstrated in vitro despite inhibition of new collagen synthesis by cycloheximide. Preservation of the collagen precursor in preparations extracted with acetic acid results from inhibition of the enzymatic activity at low pH.


Asunto(s)
Huesos/metabolismo , Colágeno/biosíntesis , Péptido Hidrolasas/metabolismo , Animales , Autorradiografía , Embrión de Pollo , Cromatografía , Colágeno/análisis , Cicloheximida/farmacología , Hueso Frontal/metabolismo , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Hueso Parietal/metabolismo , Prolina/metabolismo , Ratas , Tritio
10.
Cells Tissues Organs ; 190(3): 158-69, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19218784

RESUMEN

The mammalian skull vault consists mainly of 5 flat bones, the paired frontals and parietals, and the unpaired interparietal. All of these bones are formed by intramembranous ossification within a layer of mesenchyme, the skeletogenic membrane, located between the dermal mesenchyme and the meninges surrounding the brain. While the frontal bones are of neural crest in origin, the parietal bones arise from mesoderm. The present study is a characterization of frontal and parietal bones at their molecular level, aiming to highlight distinct differences between the neural crest-derived frontal and mesodermal-derived parietal bone. We performed a detailed comparative gene expression profile of FGF ligands and their receptors known to play crucial role in skeletogenesis. This analysis revealed that a differential expression pattern of the major FGF osteogenic molecules and their receptors exists between the neural crest-derived frontal bone and the paraxial mesoderm-derived parietal bone. Particularly, the expression of ligands such as Fgf-2, Fgf-9 and Fgf-18 was upregulated in frontal bone on embryonic day 17.5, postnatal day 1 and postnatal day 60 mice. Frontal bone also elaborated higher levels of Fgf receptor 1, 2 and 3 transcripts versus parietal bone. Taken together, these data suggest that the frontal bone is a domain with higher FGF-signaling competence than parietal bone.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Hueso Frontal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hueso Parietal/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Animales , Proliferación Celular , Células Cultivadas , Factores de Crecimiento de Fibroblastos/metabolismo , Hueso Frontal/embriología , Perfilación de la Expresión Génica , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Hueso Parietal/embriología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
11.
Clin Oral Implants Res ; 19(6): 590-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18422983

RESUMEN

OBJECTIVES: The aims of this study were to evaluate the rate of bone formation and osseointegration after topical gene delivery with a liposomal vector system carrying bone morphogenetic protein (BMP)-2 cDNA in combination with a collagen carrier and autologous bone as a carrier in freshly created peri-implant bone defects. MATERIALS AND METHODS: Eight domestic pigs received nine calvariae defects each (10 x 7 mm). A dental implant was inserted into the centre of each defect. In the test groups, the remaining space was filled with the liposomal vector/BMP-2 complex combined with a collagen carrier (n=18) or an autologous bone graft (n=18). Control groups were collagen only (n=18) and autologous bone graft only (n=18). RESULTS: There was a significant difference in mineralisation rate in the BMP-2/bone graft (29.9%+/- 4.8 and 68.3%+/- 7.2) and bone graft only (22.6%+/- 2.6 and 49.4%+/- 13.9) groups after 7 and 28 days. Mineralisation values were also significantly higher in the BMP-2/collagen group (21.2%+/- 16.2 and 53.1%+/- 12.5) compared with the collagen-only group (8.2%+/- 7 and 41%+/- 8.1) in two different regions after 28 days. Also the bone-to-implant contact was significantly increased in the BMP-2/bone graft group after 28 days and in the BMP-2/collagen group after 7 and 28 days compared with their control groups. CONCLUSIONS: The results of this study show a significantly positive effect of liposomal vector/BMP-2 on bone regeneration and osseointegration in bony circumferential peri-implant defects.


Asunto(s)
Proteínas Morfogenéticas Óseas/administración & dosificación , Regeneración Ósea/fisiología , Implantes Dentales , Hueso Frontal/metabolismo , Terapia Genética/métodos , Oseointegración/fisiología , Factor de Crecimiento Transformador beta/administración & dosificación , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/genética , Regeneración Ósea/genética , Trasplante Óseo/métodos , Trasplante Óseo/fisiología , Colágeno , ADN Complementario , Sistemas de Liberación de Medicamentos , Femenino , Regulación de la Expresión Génica/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Liposomas , Oseointegración/genética , Osteogénesis/genética , Osteogénesis/fisiología , Plásmidos/administración & dosificación , Estadísticas no Paramétricas , Sus scrofa , Factor de Crecimiento Transformador beta/genética , Trasplante Autólogo/métodos , Trasplante Autólogo/fisiología
12.
Sci Rep ; 8(1): 18021, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30575813

RESUMEN

Enlarged fontanelles and smaller frontal bones result in a mechanically compromised skull. Both phenotypes could develop from defective migration and differentiation of osteoblasts in the skull bone primordia. The Wnt/Planar cell polarity (Wnt/PCP) signaling pathway regulates cell migration and movement in other tissues and led us to test the role of Prickle1, a core component of the Wnt/PCP pathway, in the skull. For these studies, we used the missense allele of Prickle1 named Prickle1Beetlejuice (Prickle1Bj). The Prickle1Bj/Bj mutants are microcephalic and develop enlarged fontanelles between insufficient frontal bones, while the parietal bones are normal. Prickle1Bj/Bj mutants have several other craniofacial defects including a midline cleft lip, incompletely penetrant cleft palate, and decreased proximal-distal growth of the head. We observed decreased Wnt/ß-catenin and Hedgehog signaling in the frontal bone condensations of the Prickle1Bj/Bj mutants. Surprisingly, the smaller frontal bones do not result from defects in cell proliferation or death, but rather significantly delayed differentiation and decreased expression of migratory markers in the frontal bone osteoblast precursors. Our data suggests that Prickle1 protein function contributes to both the migration and differentiation of osteoblast precursors in the frontal bone.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Diferenciación Celular/genética , Hueso Frontal/embriología , Proteínas con Dominio LIM/fisiología , Osteoblastos/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Movimiento Celular/genética , Polaridad Celular/genética , Embrión de Mamíferos , Desarrollo Embrionario/genética , Células Madre Embrionarias/fisiología , Hueso Frontal/citología , Hueso Frontal/metabolismo , Proteínas con Dominio LIM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/metabolismo
13.
Mech Dev ; 152: 13-20, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29727702

RESUMEN

Protein arginine methylation has been recently identified as an important form of post-translational modification (PTM). It is carried out by the protein arginine methyltransferase (PRMT) family of enzymes, which in mammals consists of nine members. Among them, PRMT1 is the major arginine methyltransferase and participates in transcription, signal transduction, development and cancer. The function of PRMT1 in craniofacial development remains unclear. We generated Wnt1-Cre;Prmt1fl/fl mice with cranial neural crest (CNC)-specific deletion of Prmt1 and compared CNC-derived craniofacial bones from newborn control and Wnt1-Cre;Prmt1fl/fl mice. The size, surface area and volume of the premaxilla, maxilla, palatine bone, frontal bone, and mandible were analyzed using three-dimensional (3D) micro-computed tomography (microCT). We found that Prmt1 deficiency led to alterations in craniofacial bones including the premaxilla, maxilla, palatine bone, frontal bone, and mandible, as well as defects in the incisor and alveolar bone, recapitulating changes seen in Msx1-deficient mice. We further determined that Prmt1 depletion resulted in significant downregulation of Msx1 in calvaria-derived preosteoblast and primordium of frontal bone and mandible. Our study reveals critical roles of PRMT1 in the formation of CNC-derived craniofacial bones and suggests that Prmt1 is an upstream regulator of Msx1 in craniofacial bone development.


Asunto(s)
Desarrollo Óseo/genética , Factor de Transcripción MSX1/genética , Procesamiento Proteico-Postraduccional/genética , Proteína-Arginina N-Metiltransferasas/genética , Animales , Animales Modificados Genéticamente/genética , Arginina/genética , Hueso Frontal/crecimiento & desarrollo , Hueso Frontal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrasas/genética , Factor de Transcripción MSX1/deficiencia , Maxilar/crecimiento & desarrollo , Metilación , Ratones , Proteína-Arginina N-Metiltransferasas/deficiencia , Proteína Wnt1/genética
14.
PLoS One ; 11(11): e0165775, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27829009

RESUMEN

Using morphological, histological, and TEM analyses of the cranium, we provide a detailed description of bone and suture growth in zebrafish. Based on expression patterns and localization, we identified osteoblasts at different degrees of maturation. Our data confirm that, unlike in humans, zebrafish cranial sutures maintain lifelong patency to sustain skull growth. The cranial vault develops in a coordinated manner resulting in a structure that protects the brain. The zebrafish cranial roof parallels that of higher vertebrates and contains five major bones: one pair of frontal bones, one pair of parietal bones, and the supraoccipital bone. Parietal and frontal bones are formed by intramembranous ossification within a layer of mesenchyme positioned between the dermal mesenchyme and meninges surrounding the brain. The supraoccipital bone has an endochondral origin. Cranial bones are separated by connective tissue with a distinctive architecture of osteogenic cells and collagen fibrils. Here we show RNA in situ hybridization for col1a1a, col2a1a, col10a1, bglap/osteocalcin, fgfr1a, fgfr1b, fgfr2, fgfr3, foxq1, twist2, twist3, runx2a, runx2b, sp7/osterix, and spp1/ osteopontin, indicating that the expression of genes involved in suture development in mammals is preserved in zebrafish. We also present methods for examining the cranium and its sutures, which permit the study of the mechanisms involved in suture patency as well as their pathological obliteration. The model we develop has implications for the study of human disorders, including craniosynostosis, which affects 1 in 2,500 live births.


Asunto(s)
Suturas Craneales/citología , Hueso Frontal/citología , Regulación del Desarrollo de la Expresión Génica , Hueso Occipital/citología , Osteogénesis/genética , Hueso Parietal/citología , Animales , Colágeno/genética , Colágeno/metabolismo , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Suturas Craneales/crecimiento & desarrollo , Suturas Craneales/metabolismo , Hueso Frontal/crecimiento & desarrollo , Hueso Frontal/metabolismo , Humanos , Hueso Occipital/crecimiento & desarrollo , Hueso Occipital/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , Hueso Parietal/crecimiento & desarrollo , Hueso Parietal/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción Sp7 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción Twist/genética , Factores de Transcripción Twist/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
J Biomed Mater Res A ; 103(12): 3764-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26053543

RESUMEN

The skull defect model is the existing representative osteogenesis model. The skull defect model involves monitoring osteogenesis patterns at the site of a skull defect, which has the advantages that identical defects can be induced across individual experimental animals and the results can be quantitatively evaluated. However, it can damage the cerebrum because it requires a complex surgery performed on the parietal bone. This study aims to develop a new osteogenesis model that compensates for the weak points of the existing model. Male 8-week-old imprinting control region mice were put under inhalational anesthesia, and the surgery area was disinfected with 70% ethanol prior to the creation of a 5-mm incision along the sagittal line between the glabella with a pair of scissors. The incised area was opened and, after we checked the positions of the inferior cerebral vein and the sagittal suture, a 21-gauge needle was used to make two symmetrical holes with respect to the sagittal suture 3 mm below the inferior cerebral vein and 2 mm on either side of the sagittal suture. After images were obtained using micro-computed tomography, the degree of osteogenesis was quantitatively analyzed. In addition, mRNA extracted from the site of the defect confirmed a significant increase in mRNA levels of collagen 1a, alkaline phosphatase, bone sialoprotein, osteocalcin, and Runx2, known markers for osteoblasts. The promotion of osteogenesis could be observed at the site of the defect, by histological analysis.


Asunto(s)
Hueso Frontal/lesiones , Osteogénesis/efectos de los fármacos , Hormona Paratiroidea/uso terapéutico , Animales , Regeneración Ósea/efectos de los fármacos , Modelos Animales de Enfermedad , Hueso Frontal/metabolismo , Hueso Frontal/patología , Masculino , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoblastos/patología , ARN Mensajero/genética , Microtomografía por Rayos X
16.
J Bone Miner Res ; 12(10): 1664-71, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9333127

RESUMEN

Metabolic acidosis induces resorption of cultured bone, resulting in a net efflux of calcium (Ca) from the bone and an apparent loss of mineral potassium (K). However, in these organ cultures, there is diffusion of K between the medium and the crystal lattice, causing difficulty in interpretation of the acid-induced changes in mineral ion composition. To determine the effects of acidosis on bone mineral K, we injected 4-day-old neonatal mice with pure stable isotope 41K, equal to approximately 5% of their total body K. Calvariae were dissected 24 h later and then cultured for 24 h in medium without added 41K, either at pH approximately 7.4 (Ctl) or at pH approximately 7.1 (Ac), with or without the osteoclastic inhibitor calcitonin (3 x 10(-9) M, CT). The bone isotopic ion content was determined with a high-resolution scanning ion microprobe utilizing secondary ion mass spectrometry. 41K is present in nature at 6.7% of total K. The injected 41K raised the ratio of bone 41K/(39K+41K) to 9.8+/-0.5% on the surface (ratios of counts per second of detected secondary ions, mean+/-95% confidence interval) but did not alter the ratio in the interior (6.9+/-0.4%), indicating biological incorporation of the 41K into the mineral surface. The ratios of 41K/40Ca on the surface of Ctl calvariae was 14.4+/-1.2, indicating that bone mineral surface is rich in K compared with Ca. Compared with Ctl, Ac caused a marked increase in the net Ca efflux from bone that was blocked by CT. Ac also induced a marked fall in the ratio of 41K/40Ca on the surface of the calvariae (43+/-0.5, p < 0.01 vs. Ctl), which was partially blocked by CT (8.2+/-0.9, p < 0.01 vs. Ctl and vs. Ac), indicating that Ac causes a greater release of bone mineral K than Ca which is partially blocked by CT. Thus, bone mineral surface is rich in K relative to Ca, acidosis induces a greater release of surface mineral K than Ca, and osteoclastic function is necessary to support the enriched levels of surface mineral K in the presence of acidosis.


Asunto(s)
Acidosis/fisiopatología , Potasio/metabolismo , Cráneo/metabolismo , Animales , Animales Recién Nacidos , Densidad Ósea , Calcitonina/farmacología , Calcio/metabolismo , Hueso Frontal/metabolismo , Hueso Frontal/ultraestructura , Concentración de Iones de Hidrógeno , Marcaje Isotópico , Espectrometría de Masas , Ratones , Microscopía Electrónica de Rastreo , Técnicas de Cultivo de Órganos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoclastos/ultraestructura , Hueso Parietal/metabolismo , Hueso Parietal/ultraestructura , Isótopos de Potasio
17.
J Bone Miner Res ; 15(3): 522-33, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10750567

RESUMEN

Calvarial and facial bones form by intramembranous ossification, in which bone cells arise directly from mesenchyme without an intermediate cartilage anlage. However, a number of studies have reported the emergence of chondrocytes from in vitro calvarial cell or organ cultures and the expression of type II collagen, a cartilage-characteristic marker, in developing calvarial bones. Based on these findings we hypothesized that a covert chondrogenic phase may be an integral part of the normal intramembranous pathway. To test this hypothesis, we analyzed the temporal and spatial expression patterns of cartilage characteristic genes in normal membranous bones from chick embryos at various developmental stages (days 12, 15 and 19). Northern and RNAse protection analyses revealed that embryonic frontal bones expressed not only the type I collagen gene but also a subset of cartilage characteristic genes, types IIA and XI collagen and aggrecan, thus resembling a phenotype of prechondrogenic-condensing mesenchyme. The expression of cartilage-characteristic genes decreased with the progression of bone maturation. Immunohistochemical analyses of developing embryonic chick heads indicated that type II collagen and aggrecan were produced by alkaline phosphatase activity positive cells engaged in early stages of osteogenic differentiation, such as cells in preosteogenic-condensing mesenchyme, the cambium layer of periosteum, the advancing osteogenic front, and osteoid bone. Type IIB and X collagen messenger RNAs (mRNA), markers for mature chondrocytes, were also detected at low levels in calvarial bone but not until late embryonic stages (day 19), indicating that some calvarial cells may undergo overt chondrogenesis. On the basis of our findings, we propose that the normal intramembranous pathway in chicks includes a previously unrecognized transient chondrogenic phase similar to prechondrogenic mesenchyme, and that the cells in this phase retain chondrogenic potential that can be expressed in specific in vitro and in vivo microenvironments.


Asunto(s)
Cartílago/embriología , Proteínas de la Matriz Extracelular , Hueso Frontal/embriología , Osteogénesis/fisiología , Agrecanos , Fosfatasa Alcalina/análisis , Animales , Biomarcadores , Cartílago/citología , Cartílago/metabolismo , Diferenciación Celular , Linaje de la Célula , Embrión de Pollo , Colágeno/biosíntesis , Colágeno/genética , Hueso Frontal/citología , Hueso Frontal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Lectinas Tipo C , Mesodermo/citología , Osteoblastos/metabolismo , Procolágeno/biosíntesis , Procolágeno/genética , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Proteoglicanos/biosíntesis , Proteoglicanos/genética , ARN Mensajero/biosíntesis , Cráneo/citología , Cráneo/embriología , Cráneo/metabolismo , Esternón/embriología , Esternón/metabolismo
18.
Neurobiol Aging ; 25(1): 71-81, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14675733

RESUMEN

Oxidative damage to mitochondrial DNA (mtDNA) increases with age in the brain and can induce G:C to T:A and T:A to G:C point mutations. Though rare at any particular site, multiple somatic mtDNA mutations induced by oxidative damage or by other mechanisms may accumulate with age in the brain and thus could play a role in aging and neurodegenerative diseases. However, no prior study has quantified the total burden of mtDNA point mutation subtypes in the brain. Using a highly sensitive cloning and sequencing strategy, we find that the aggregate levels of G:C to T:A and T:A to G:C transversions and of all point mutations increase with age in the frontal cortex (FCtx). In the substantia nigra (SN), the aggregate levels of point mutations in young controls are similar to the levels in the SN or FCtx of elderly subjects. Extrapolation from our data suggests an average of 2.7 (FCtx) to 3.2 (SN) somatic point mutations per mitochondrial genome in elderly subjects. There were no significant differences between Parkinson's disease (PD) patients and age-matched controls in somatic mutation levels. These results indicate that individually rare mtDNA point mutations reach a high aggregate burden in FCtx and SN of elderly subjects.


Asunto(s)
Envejecimiento/genética , ADN Mitocondrial/genética , Hueso Frontal/patología , Enfermedad de Parkinson/genética , Mutación Puntual , Sustancia Negra/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Línea Celular/metabolismo , Niño , Preescolar , Análisis Mutacional de ADN , ADN Mitocondrial/metabolismo , Femenino , Hueso Frontal/metabolismo , Humanos , Lactante , Masculino , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , Sustancia Negra/metabolismo
19.
Br J Oral Maxillofac Surg ; 52(4): 308-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24582013

RESUMEN

Osteonecrosis of the jaw as a result of treatment with receptor activators of nuclear factor kappa-B ligand (RANKL) inhibitors (denosumab) is a new type of bony necrosis, the exact pathogenesis of which is unknown. Our aim was to find out whether the turnover of bone in the jaw is increased after denosumab has been given compared with other skeletal sites, and if that turnover might have a role in denosumab-related osteonecrosis of the jaw (DRONJ). Bone scintigraphic images of 45 female patients with breast cancer and bone metastases were analysed retrospectively, and divided into 3 groups: those given denosumab, those given a bisphosphonate, and a control group (n=15 in each). All patients had bone scintigraphy before treatment (T0) and during the course of treatment after 12 (T1) and 24 (T2) months. The data were analysed quantitatively using 6 preset bony regions of interest. There was similar turnover of bone in the mandible compared with other skeletal sites (such as the femur), while the maxilla showed significantly higher turnover. None of the bony regions investigated showed any significant changes after the bisphosphonate had been given. There was a tendency to increase bone turnover in those patients taking denosumab. The bone turnover of the jawbone is not overtly changed either by a bisphosphonate or denosumab, so it seems unlikely that oversuppression of bony turnover in the jawbones plays an important part either in the pathogenesis of DRONJ or in the bisphosphonate-related osteonecrosis of the jaw (BRONJ).


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Remodelación Ósea/efectos de los fármacos , Difosfonatos/uso terapéutico , Mandíbula/efectos de los fármacos , Maxilar/efectos de los fármacos , Ligando RANK/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Osteonecrosis de los Maxilares Asociada a Difosfonatos/etiología , Osteonecrosis de los Maxilares Asociada a Difosfonatos/metabolismo , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/secundario , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/diagnóstico por imagen , Carcinoma/tratamiento farmacológico , Carcinoma/secundario , Denosumab , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/metabolismo , Hueso Frontal/diagnóstico por imagen , Hueso Frontal/efectos de los fármacos , Hueso Frontal/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Imidazoles/uso terapéutico , Enfermedades Maxilomandibulares/inducido químicamente , Enfermedades Maxilomandibulares/metabolismo , Mandíbula/diagnóstico por imagen , Mandíbula/metabolismo , Maxilar/diagnóstico por imagen , Maxilar/metabolismo , Persona de Mediana Edad , Osteonecrosis/inducido químicamente , Osteonecrosis/metabolismo , Cintigrafía , Estudios Retrospectivos , Ácido Zoledrónico
20.
PLoS One ; 7(5): e36789, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693558

RESUMEN

Growth Differentiation Factor-6 (Gdf6) is a member of the Bone Morphogenetic Protein (BMP) family of secreted signaling molecules. Previous studies have shown that Gdf6 plays a role in formation of a diverse subset of skeletal joints. In mice, loss of Gdf6 results in fusion of the coronal suture, the intramembranous joint that separates the frontal and parietal bones. Although the role of GDFs in the development of cartilaginous limb joints has been studied, limb joints are developmentally quite distinct from cranial sutures and how Gdf6 controls suture formation has remained unclear. In this study we show that coronal suture fusion in the Gdf6-/- mouse is due to accelerated differentiation of suture mesenchyme, prior to the onset of calvarial ossification. Gdf6 is expressed in the mouse frontal bone primordia from embryonic day (E) 10.5 through 12.5. In the Gdf6-/- embryo, the coronal suture fuses prematurely and concurrently with the initiation of osteogenesis in the cranial bones. Alkaline phosphatase (ALP) activity and Runx2 expression assays both showed that the suture width is reduced in Gdf6+/- embryos and is completely absent in Gdf6-/- embryos by E12.5. ALP activity is also increased in the suture mesenchyme of Gdf6+/- embryos compared to wild-type. This suggests Gdf6 delays differentiation of the mesenchyme occupying the suture, prior to the onset of ossification. Therefore, although BMPs are known to promote bone formation, Gdf6 plays an inhibitory role to prevent the osteogenic differentiation of the coronal suture mesenchyme.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Suturas Craneales/embriología , Factor 6 de Diferenciación de Crecimiento/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Suturas Craneales/citología , Suturas Craneales/metabolismo , Hueso Frontal/citología , Hueso Frontal/embriología , Hueso Frontal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor 6 de Diferenciación de Crecimiento/deficiencia , Ligandos , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda