Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Genesis ; 49(10): 784-96, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21557453

ABSTRACT

Much is known regarding the role of Indian hedgehog (Ihh) in endochondral ossification, where Ihh regulates multiple steps of chondrocyte differentiation. The Ihh-/- phenotype is most notable for severely foreshortened limbs and a complete absence of mature osteoblasts. A far less explored phenotype in the Ihh-/- mutant is found in the calvaria, where bones form predominately through intramembranous ossification. We investigated the role of Ihh in calvarial bone ossification, finding that proliferation was largely unaffected. Instead, our results indicate that Ihh is a pro-osteogenic factor that positively regulates intramembranous ossification. We confirmed through histologic and quantitative gene analysis that loss of Ihh results in reduction of cranial bone size and all markers of osteodifferentiation. Moreover, in vitro studies suggest that Ihh loss reduces Bmp expression within the calvaria, an observation that may underlie the Ihh-/- calvarial phenotype. In conjunction with the newly recognized roles of Hedgehog deregulation in craniosynostosis, our study defines Ihh as an important positive regulator of cranial bone ossification.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Hedgehog Proteins/metabolism , Osteogenesis , Signal Transduction , Skull/embryology , Animals , Bone Development/genetics , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Bone and Bones/embryology , Cell Differentiation , Chondrocytes/metabolism , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism
2.
Cells Tissues Organs ; 190(6): 336-46, 2009.
Article in English | MEDLINE | ID: mdl-19590164

ABSTRACT

BACKGROUND/AIMS: Craniosynostosis, the premature fusion of cranial sutures, is a common congenital defect. In vivo models for studying cranial suture biology impose inherent restrictions on tissue accessibility and manipulation. The present study was performed to investigate the utility of the renal capsule assay in overcoming these limitations and providing a reproducible model system for studying cranial suture morphogenesis and fate. MATERIALS AND METHODS: The posterior frontal suture, which fuses physiologically, and the coronal and sagittal sutures, which remain patent, were dissected from postnatal and embryonic mouse calvaria and placed under the renal capsule of syngeneic recipient mice (n = 72 in total). Sutures were harvested from 1-14 days after transplantation for histological and morphometric analysis. Suture transplants were compared with nonmanipulated sutures at equivalent developmental stages. The derivation of cells associated with the growing transplants was analyzed using beta-actin-GFP (green fluorescent protein) transgenic mice. RESULTS: Sutures transplanted under the renal capsule maintained normal suture morphology and fate with the posterior frontal suture fusing and the coronal and sagittal sutures remaining patent. In posterior frontal suture transplants, the fusion process mimicked in vivo suture fusion with a delay of 1-2 days. In comparison to in vivo suture complexes, transplant thickness and trabeculation were significantly increased. In addition, we found that osteoblasts within the growing transplant were derived from the transplant itself rather than the host. CONCLUSION: The renal capsule supports the growth of cranial sutures. In this system transplanted sutures recapitulate the anatomical development and fate (fusion or patency) of cranial sutures in vivo. This model system will facilitate controlled ex vivo manipulations of both embryonic and postnatal sutures.


Subject(s)
Cranial Sutures/growth & development , Morphogenesis , Subrenal Capsule Assay/methods , Animals , Cranial Sutures/cytology , Cranial Sutures/transplantation , Craniosynostoses , Disease Models, Animal , In Vitro Techniques , Mice , Osteoblasts/cytology , Osteoblasts/physiology
3.
Plast Reconstr Surg ; 121(4): 170e-178e, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18349596

ABSTRACT

Craniosynostosis, or the premature fusion of one or more cranial sutures, is a relatively common congenital defect that causes a number of morphologic and functional abnormalities. With advances in genetics and molecular biology, research of craniosynostosis has progressed from describing gross abnormalities to understanding the molecular interactions that underlie these cranial deformities. Animal models have been extremely valuable in improving our comprehension of human craniofacial morphogenesis, primarily by human genetic linkage analysis and the development of knock-out animals. This article provides a brief review of perisutural tissue interactions, embryonic origins, signaling molecules and their receptors, and transcription factors in maintaining the delicate balance between proliferation and differentiation of cells within the suture complex that determines suture fate. Finally, this article discusses the potential implications for developing novel therapies for craniosynostosis.


Subject(s)
Craniosynostoses/embryology , Suture Techniques , Animals , Disease Models, Animal , Humans , Skull/embryology
4.
Plast Reconstr Surg ; 116(5): 1353-62, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16217479

ABSTRACT

BACKGROUND: Clinical genetics data and investigative studies have contributed greatly to our understanding of the role of numerous genes in craniosynostosis. Recent studies have introduced antagonists of osteogenesis as potential key regulators of suture fusion and patency. The authors investigated the expression pattern of the bone morphogenetic protein antagonist BMP3 in rat cranial sutures and the factors regulating its expression in vitro. METHODS: Microarray analysis was performed on rat posterior frontal and sagittal cranial sutures at 5, 10, 15, 20, and 30 days of life (n = 30 per group). Gene expression was confirmed using quantitative real-time reverse transcriptase polymerase chain reaction. Regulation of BMP3 expression was determined using primary rat calvarial osteoblasts stimulated with recombinant human fibroblast growth factor 2 or recombinant human transforming growth factor beta1, or cultured with primary rat nonsuture dura mater. Gene expression was quantified with quantitative real-time reverse transcriptase polymerase chain reaction. RESULTS: BMP3 expression in the posterior frontal suture decreased over the time course analyzed, whereas it increased in the sagittal suture. Notably, BMP3 expression was higher in the patent sagittal suture during the window of posterior frontal suture fusion. Stimulation of osteoblasts with recombinant human fibroblast growth factor 2 led to a rapid and sustained suppression of BMP3 expression (85 percent, p < 0.01) when compared with controls. Co-culture with dural cells decreased BMP3 mRNA by 50 percent compared with controls (p < 0.01). CONCLUSIONS: BMP3 is expressed in rat cranial sutures in a temporal pattern suggesting involvement in cranial suture patency and fusion. Furthermore, BMP3 is regulated in calvarial osteoblasts by recombinant human fibroblast growth factor 2 and by paracrine signaling from dura mater. These data add to our knowledge of the role of osteogenic antagonists in cranial suture biology.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cranial Sutures/metabolism , Oligonucleotide Array Sequence Analysis , Osteogenesis/physiology , Transforming Growth Factor beta/pharmacology , Animals , Bone Morphogenetic Protein 3 , Carrier Proteins , Cells, Cultured , Coculture Techniques , Down-Regulation/physiology , Dura Mater/cytology , Fibroblast Growth Factor 2/pharmacology , Osteoblasts/cytology , Osteoblasts/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1
5.
Plast Reconstr Surg ; 115(7): 1986-94, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923847

ABSTRACT

BACKGROUND: It has widely been observed that young children are capable of reossifying large calvarial defects, while adults lack this endogenous tissue-engineering capacity. The ability of juvenile animals to regenerate calvarial defects has been investigated in multiple animal models, including mice. In this study, the authors used cDNA microarrays to investigate the expression of osteogenesis-associated genes upstream and downstream of Runx2 in juvenile and adult mouse calvaria. METHODS: Nonsuture-associated parietal bone discs were harvested from 6-day-old (n = 50) and 60-day-old (n = 35) male CD-1 mice. After separation of the underlying dura mater and overlying pericranium, the calvarial discs were snap-frozen and RNA was extracted from pooled samples of calvaria for microarray analysis. Genes analyzed included cytokines, receptors, and cell-surface and matrix proteins both upstream and downstream of Runx2. RESULTS: Genes associated with the Runx2 pathway had notably higher levels in the juvenile versus adult calvaria. All genes except for osteocalcin were expressed at least twofold higher in the juvenile calvaria. This pattern was validated with quantitative real-time polymerase chain reaction. In addition, mRNA for potent osteoinductive growth factors was present at higher levels in the juvenile compared with the adult calvaria. CONCLUSIONS: These findings reflect a genomic environment of active osteoblast differentiation and ossification in the juvenile calvaria compared with the adult "quiescent" calvarial tissue. These data suggest that a decreased osteogenic potential of adult calvarial osteoblasts may, in part, explain the inability of adult animals to heal calvarial defects.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Osteogenesis/genetics , Skull/metabolism , Age Factors , Animals , Cell Differentiation/genetics , Extracellular Matrix Proteins/metabolism , Male , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
7.
Plast Reconstr Surg ; 115(3): 838-52, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731686

ABSTRACT

An abundance of genetic and experimental data have suggested that fibroblast growth factor (FGF) signaling plays a central role in physiological and pathological cranial suture fusion. Although alterations in the differentiation and proliferation of sutural osteoblasts may be a key mediator of this process, the mechanisms by which FGF signaling regulates osteoblast differentiation remain incompletely understood. In the current study, the authors show that recombinant human FGF-2 alters osteoblastic expression of bone morphogenetic protein-2 and Msx-2 in vitro to favor cellular differentiation and osteoinduction. The ERK1/2 intracellular signaling cascade was shown to be necessary for recombinant human FGF-2-mediated bone morphogenetic protein-2 transcriptional changes. Furthermore, the cellular production of an intermediate transcriptional modifier was found to be necessary for the recombinant human FGF-2-mediated gene expression changes in bone morphogenetic protein-2 and Msx-2. Together, these findings offer new insight into the mechanisms by which FGF-2 modulates osteoblast biology.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Fibroblast Growth Factor 2/physiology , Homeodomain Proteins/physiology , Osteoblasts/physiology , Transforming Growth Factor beta/metabolism , Animals , Animals, Newborn , Blotting, Northern , Bone Morphogenetic Protein 2 , Gene Expression , Immunoblotting , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Skull/cytology , Up-Regulation/physiology
8.
Clin Plast Surg ; 32(1): 123-36, ix-x, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15636770

ABSTRACT

Pediatric plastic surgery research is a rapidly expanding field. Unique in many ways, researchers in this field stand at the union of multiple scientific specialties, including biomedical engineering, tissue engineering, polymer science, molecular biology, developmental biology, and genetics. The goal of this scientific effort is to translate research advances into improved treatments for children with congenital and acquired defects. Although the last decade has seen a dramatic acceleration in research related to pediatric plastic surgery, the next 10 years will no doubt lead to novel treatment strategies with improved clinical outcomes.


Subject(s)
Cranial Sutures/physiology , Osteogenesis, Distraction , Plastic Surgery Procedures , Tissue Engineering , Animals , Biomedical Research , Child , Humans
9.
Plast Reconstr Surg ; 114(3): 713-20, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15318051

ABSTRACT

Young children are capable of healing large calvarial defects, whereas adults lack this endogenous osseous tissue-engineering capacity. Despite the important clinical implications, little is known about the molecular and cell biology underlying this differential ability. Traditionally, guinea pig, rabbit, and rat models have been used to study the orchestration of calvarial healing. To harness the research potential of knockout and transgenic mice, the authors developed a mouse model for calvarial healing. Nonsuture-associated parietal defects 3, 4, and 5 mm in diameter were made in both juvenile (6-day-old, n = 15) and adult (60-day-old, n = 15) mice. Calvariae were harvested after 8 weeks and analyzed radiographically and histologically. Percentage of healing was quantified using Scion Image software analysis of calvarial radiographs. A significant difference in the ability to heal calvarial defects was seen between 6-day-old and 60-day-old mice when 3-, 4-, or 5-mm defects were created. The authors' analysis revealed that juvenile mice healed a significantly greater percentage of their calvarial defects than adult mice (juvenile mean percentage of healing: 3-mm defects, 59 percent; 4-mm defects, 65 percent; 5-mm defects, 44 percent; adult mean percentage of healing: <5 percent in all groups; p < 0.05). All three defect sizes were found to be critical in the adult, whereas significant healing was seen regardless of the size of the defect in juvenile mice. The establishment of this model will facilitate further, detailed evaluation of the molecular biology underlying the different regenerative abilities of juvenile versus adult mice and enhance research into membranous bone induction by making available powerful tools such as knockout and transgenic animals.


Subject(s)
Bone Regeneration/physiology , Models, Animal , Models, Biological , Skull/physiology , Age Factors , Animals , Mice , Mice, Inbred Strains , Osteogenesis/physiology , Radiography , Skull/diagnostic imaging , Skull/pathology , Wound Healing/physiology
11.
Plast Reconstr Surg ; 112(2): 528-39, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12900611

ABSTRACT

Appropriately timed closure of the cranial sutures is a critical factor in normal postnatal morphogenesis of the cranial vault. Suture patency is necessary to permit rapid neonatal expansion of the cerebral hemispheres, and later ossification is important for bony protection of the cerebrum. Premature suture ossification (craniosynostosis) leads to myriad adverse functional and developmental consequences. Several murine studies have implicated dura-derived fibroblast growth factor-2 (FGF-2) paracrine signaling as a critical factor promoting physiologic posterior frontal suture fusion. In this study, the authors used real-time reverse transcription polymerase chain reaction (RT-PCR) to study an in vitro system that models the in vivo stimulation of suture calvarial osteoblasts by dura-derived FGF-2. The authors advocate real-time RT-PCR as a powerful and rapid technique that offers advantages in the highly sensitive, specific, and reproducible analyses of nine genes known to be important in cranial suture biology. The genes studied were growth factors [FGF-2, transforming growth factor (TGF)-beta 1, TGF-beta 2, and TGF-beta 3], growth factor receptors (FGF-R1, FGF-R2, TGF-beta RI, and TGF-beta RII), and a marker of osteoblast differentiation (Co1-I alpha I). These analyses provide a "snapshot" of several important genes involved in suture fusion that is more inclusive and quantitative than that which has been previously reported.


Subject(s)
Cranial Sutures/physiology , Fibroblast Growth Factor 2/physiology , Osteoblasts/physiology , Receptors, Fibroblast Growth Factor/genetics , Receptors, Transforming Growth Factor beta/genetics , Skull/cytology , Transforming Growth Factor beta/genetics , Animals , Animals, Newborn , Cells, Cultured , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Mice , Paracrine Communication , RNA, Messenger/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...