Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells Dev ; 32(13-14): 398-409, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37078151

RESUMO

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.


Assuntos
Suturas Cranianas , Osso Frontal , Animais , Feminino , Masculino , Camundongos , Suturas Cranianas/metabolismo , Osso Frontal/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Histonas/metabolismo , Camundongos Knockout , Osteogênese/genética , Fatores Sexuais
2.
Stem Cell Res Ther ; 13(1): 341, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35883153

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Células-Tronco Mesenquimais , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Ectoderma/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Osso Frontal/metabolismo , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Ácido Glutâmico/uso terapêutico , Inflamação/metabolismo , Inflamação/terapia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neuroinflamatórias , Osteogênese
3.
Dev Dyn ; 248(10): 1009-1019, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31397024

RESUMO

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.


Assuntos
Osso Frontal/metabolismo , MicroRNAs/análise , Osso Parietal/metabolismo , Crânio/metabolismo , Animais , Regeneração Óssea , Adesões Focais , Insulina/metabolismo , Camundongos , MicroRNAs/fisiologia , Osteogênese , Transdução de Sinais
4.
Med Mol Morphol ; 52(3): 156-163, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30499042

RESUMO

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.


Assuntos
Condrogênese , Osso Frontal/crescimento & desenvolvimento , Crista Neural/crescimento & desenvolvimento , Osteogênese , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Osso Frontal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Crista Neural/citologia , Crista Neural/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética
5.
Sci Rep ; 8(1): 18021, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575813

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Diferenciação Celular/genética , Osso Frontal/embriologia , Proteínas com Domínio LIM/fisiologia , Osteoblastos/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Movimento Celular/genética , Polaridade Celular/genética , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/fisiologia , Osso Frontal/citologia , Osso Frontal/metabolismo , Proteínas com Domínio LIM/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/metabolismo
6.
Mech Dev ; 152: 13-20, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29727702

RESUMO

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.


Assuntos
Desenvolvimento Ósseo/genética , Fator de Transcrição MSX1/genética , Processamento de Proteína Pós-Traducional/genética , Proteína-Arginina N-Metiltransferases/genética , Animais , Animais Geneticamente Modificados/genética , Arginina/genética , Osso Frontal/crescimento & desenvolvimento , Osso Frontal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Integrases/genética , Fator de Transcrição MSX1/deficiência , Maxila/crescimento & desenvolvimento , Metilação , Camundongos , Proteína-Arginina N-Metiltransferases/deficiência , Proteína Wnt1/genética
7.
PLoS One ; 11(11): e0165775, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27829009

RESUMO

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.


Assuntos
Suturas Cranianas/citologia , Osso Frontal/citologia , Regulação da Expressão Gênica no Desenvolvimento , Osso Occipital/citologia , Osteogênese/genética , Osso Parietal/citologia , Animais , Colágeno/genética , Colágeno/metabolismo , Subunidades alfa de Fatores de Ligação ao Core/genética , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Suturas Cranianas/crescimento & desenvolvimento , Suturas Cranianas/metabolismo , Osso Frontal/crescimento & desenvolvimento , Osso Frontal/metabolismo , Humanos , Osso Occipital/crescimento & desenvolvimento , Osso Occipital/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , Osso Parietal/crescimento & desenvolvimento , Osso Parietal/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição Twist/genética , Fatores de Transcrição Twist/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
BMC Dev Biol ; 16(1): 37, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27756203

RESUMO

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.


Assuntos
Craniossinostoses/genética , Proteínas de Ligação a DNA/genética , Osso Frontal/metabolismo , Osso Parietal/metabolismo , Fatores de Transcrição/genética , Animais , Suturas Cranianas/anormalidades , Suturas Cranianas/metabolismo , Craniossinostoses/embriologia , Craniossinostoses/metabolismo , Proteínas de Ligação a DNA/deficiência , Desenvolvimento Embrionário/genética , Osso Frontal/anormalidades , Osso Frontal/diagnóstico por imagem , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos Knockout , Osso Parietal/anormalidades , Osso Parietal/diagnóstico por imagem , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Fatores de Risco , Crânio/anormalidades , Crânio/metabolismo , Fatores de Transcrição/deficiência , Microtomografia por Raio-X
9.
J Biomed Mater Res A ; 103(12): 3764-71, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26053543

RESUMO

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.


Assuntos
Osso Frontal/lesões , Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/uso terapêutico , Animais , Regeneração Óssea/efeitos dos fármacos , Modelos Animais de Doenças , Osso Frontal/metabolismo , Osso Frontal/patologia , Masculino , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoblastos/patologia , RNA Mensageiro/genética , Microtomografia por Raio-X
11.
Br J Oral Maxillofac Surg ; 52(4): 308-13, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24582013

RESUMO

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).


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Remodelação Óssea/efeitos dos fármacos , Difosfonatos/uso terapêutico , Mandíbula/efeitos dos fármacos , Maxila/efeitos dos fármacos , Ligante RANK/antagonistas & inibidores , Adulto , Idoso , Idoso de 80 Anos ou mais , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/etiologia , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/metabolismo , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/secundário , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Carcinoma/diagnóstico por imagem , Carcinoma/tratamento farmacológico , Carcinoma/secundário , Denosumab , Feminino , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Osso Frontal/diagnóstico por imagem , Osso Frontal/efeitos dos fármacos , Osso Frontal/metabolismo , Humanos , Processamento de Imagem Assistida por Computador/métodos , Imidazóis/uso terapêutico , Doenças Maxilomandibulares/induzido quimicamente , Doenças Maxilomandibulares/metabolismo , Mandíbula/diagnóstico por imagem , Mandíbula/metabolismo , Maxila/diagnóstico por imagem , Maxila/metabolismo , Pessoa de Meia-Idade , Osteonecrose/induzido quimicamente , Osteonecrose/metabolismo , Cintilografia , Estudos Retrospectivos , Ácido Zoledrônico
12.
Development ; 140(5): 1034-44, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23344708

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Osso Frontal/embriologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Animais , Desenvolvimento Ósseo/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Proteínas Morfogenéticas Ósseas/farmacologia , Diferenciação Celular/genética , Células Cultivadas , Embrião de Mamíferos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Osso Frontal/crescimento & desenvolvimento , Osso Frontal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteogênese/genética , Osteogênese/fisiologia , Crânio/embriologia , Crânio/metabolismo
13.
PLoS One ; 7(5): e36789, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22693558

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular , Suturas Cranianas/embriologia , Fator 6 de Diferenciação de Crescimento/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Suturas Cranianas/citologia , Suturas Cranianas/metabolismo , Osso Frontal/citologia , Osso Frontal/embriologia , Osso Frontal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator 6 de Diferenciação de Crescimento/deficiência , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Osteogênese , Fatores de Tempo
14.
Genesis ; 48(11): 645-55, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20824629

RESUMO

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.


Assuntos
Osso Frontal/embriologia , Proteínas de Homeodomínio/fisiologia , Fator de Transcrição MSX1/fisiologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos , Epistasia Genética/fisiologia , Osso Frontal/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fator de Transcrição MSX1/genética , Fator de Transcrição MSX1/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Knockout , Morfogênese/genética , Morfogênese/fisiologia , Osteogênese/genética , Osteogênese/fisiologia
16.
Dev Biol ; 331(2): 176-88, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414008

RESUMO

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.


Assuntos
Morfogênese/fisiologia , Crista Neural/embriologia , Proteínas Nucleares/fisiologia , Crânio/embriologia , Proteína 1 Relacionada a Twist/fisiologia , Animais , Região Branquial/citologia , Região Branquial/embriologia , Região Branquial/fisiologia , Osso Frontal/embriologia , Osso Frontal/metabolismo , Arcada Osseodentária/embriologia , Arcada Osseodentária/metabolismo , Camundongos , Camundongos Mutantes , Osso Nasal/embriologia , Osso Nasal/metabolismo , Crista Neural/citologia , Crista Neural/fisiologia , Crânio/citologia , Crânio/fisiologia
17.
Development ; 136(5): 855-64, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19201948

RESUMO

Heterozygous loss of Twist1 function causes coronal synostosis in both mice and humans. We showed previously that in mice this phenotype is associated with a defect in the neural crest-mesoderm boundary within the coronal suture, as well as with a reduction in the expression of ephrin A2 (Efna2), ephrin A4 (Efna4) and EphA4 in the coronal suture. We also demonstrated that mutations in human EFNA4 are a cause of non-syndromic coronal synostosis. Here we investigate the cellular mechanisms by which Twist1, acting through Eph-ephrin signaling, regulates coronal suture development. We show that EphA4 mutant mice exhibit defects in the coronal suture and neural crest-mesoderm boundary that phenocopy those of Twist1(+/-) mice. Further, we demonstrate that Twist1 and EphA4 interact genetically: EphA4 expression in the coronal suture is reduced in Twist1 mutants, and compound Twist1-EphA4 heterozygotes have suture defects of greater severity than those of individual heterozygotes. Thus, EphA4 is a Twist1 effector in coronal suture development. Finally, by DiI labeling of migratory osteogenic precursor cells that contribute to the frontal and parietal bones, we show that Twist1 and EphA4 are required for the exclusion of such cells from the coronal suture. We suggest that the failure of this process in Twist1 and EphA4 mutants is the cause of craniosynostosis.


Assuntos
Craniossinostoses/embriologia , Craniossinostoses/metabolismo , Proteínas Nucleares/metabolismo , Osteogênese/fisiologia , Receptor EphA4/metabolismo , Crânio/embriologia , Crânio/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Movimento Celular , Suturas Cranianas/embriologia , Suturas Cranianas/metabolismo , Craniossinostoses/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Osso Frontal/embriologia , Osso Frontal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Crista Neural/embriologia , Crista Neural/metabolismo , Proteínas Nucleares/genética , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , Osso Parietal/embriologia , Osso Parietal/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor EphA4/genética , Crânio/citologia , Proteína 1 Relacionada a Twist/genética
18.
Cells Tissues Organs ; 190(3): 158-69, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19218784

RESUMO

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.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Osso Frontal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Osso Parietal/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Animais , Proliferação de Células , Células Cultivadas , Fatores de Crescimento de Fibroblastos/metabolismo , Osso Frontal/embriologia , Perfilação da Expressão Gênica , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese , Osso Parietal/embriologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
19.
Clin Oral Implants Res ; 19(6): 590-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18422983

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/administração & dosagem , Regeneração Óssea/fisiologia , Implantes Dentários , Osso Frontal/metabolismo , Terapia Genética/métodos , Osseointegração/fisiologia , Fator de Crescimento Transformador beta/administração & dosagem , Animais , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/genética , Regeneração Óssea/genética , Transplante Ósseo/métodos , Transplante Ósseo/fisiologia , Colágeno , DNA Complementar , Sistemas de Liberação de Medicamentos , Feminino , Regulação da Expressão Gênica/fisiologia , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Lipossomos , Osseointegração/genética , Osteogênese/genética , Osteogênese/fisiologia , Plasmídeos/administração & dosagem , Estatísticas não Paramétricas , Sus scrofa , Fator de Crescimento Transformador beta/genética , Transplante Autólogo/métodos , Transplante Autólogo/fisiologia
20.
Development ; 133(2): 371-81, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16368934

RESUMO

The murine frontal bone derives entirely from the cranial neural crest (CNC) and consists of the calvarial (lateral) aspect that covers the frontal lobe of brain and the orbital aspect that forms the roof of bony orbit. TGFbeta and FGF signaling have important regulatory roles in postnatal calvarial development. Our previous study has demonstrated that conditional inactivation of Tgfbr2 in the neural crest results in severe defects in calvarial development, although the cellular and molecular mechanisms by which TGFbeta signaling regulates the fate of CNC cells during frontal bone development remain unknown. Here, we show that TGFbeta IIR is required for proliferation of osteoprogenitor cells in the CNC-derived frontal bone anlagen. FGF acts downstream of TGFbeta signaling in regulating CNC cell proliferation, and exogenous FGF2 rescues the cell proliferation defect in the frontal primordium of Tgfbr2 mutant. Furthermore, the CNC-derived frontal primordium requires TGFbeta IIR to undergo terminal differentiation. However, this requirement is restricted to the developing calvarial aspect of the frontal bone, whereas the orbital aspect forms despite the ablation of Tgfbr2 gene, implying a differential requirement for TGFbeta signaling during the development of various regions of the frontal bone. This study demonstrates the biological significance of TGFbeta-mediated FGF signaling cascade in regulating frontal bone development, suggests that TGFbeta functions as a morphogen in regulating the fate of the CNC-derived osteoblast and provides a model for investigating abnormal craniofacial development.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Osso Frontal/embriologia , Crista Neural/citologia , Crista Neural/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Modelos Animais de Doenças , Feminino , Osso Frontal/citologia , Osso Frontal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Gravidez , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...