Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Orthod Craniofac Res ; 20 Suppl 1: 12-17, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28643920

ABSTRACT

OBJECTIVES: Recurrence of cranial bone fusion following surgical resection in craniosynostosis patients commonly requires additional surgical procedures. Surgical implantation of engineered 3D scaffolds that control tissue mineralization could be utilized to diminish recurrence of fusion. This study investigated the ability of composite scaffolds to control tissue mineralization when cultured in vitro. SETTING AND SAMPLE POPULATION: Precision-engineered scaffolds with calvarial cells were cultured in vitro at the Department of Orthodontics and Pediatric Dentistry, University of Michigan. MATERIAL & METHODS: Polycaprolactone (PCL) scaffolds were fabricated using a novel precision extrusion deposition technique. Polyethylene glycol (PEG) hydrogel was coated onto select scaffolds to inhibit mineralization. MC3T3E1(C4) calvarial cells were cultured with scaffolds in media containing ascorbate and phosphate to promote osteoblast differentiation and mineralization. Scaffolds were assayed for osteoblast differentiation by alkaline phosphatase assay. Scaffolds were assayed for mineralization by nano-computed tomography (nano-CT) and by von Kossa staining of histologic sections. RESULTS: MC3T3E1(C4) cells differentiated into osteoblasts and formed mineral when cultured on uncoated PCL scaffolds. MC3T3E1(C4) cells were significantly diminished in their ability to differentiate into osteoblasts when cultured on hydrogel-coated scaffold. CONCLUSION: Results of this study indicate that this novel printing technology can be used to fabricate 3D scaffolds to promote and inhibit tissue mineralization in a region-specific manner. Future studies are needed to establish utility of such scaffolds in vivo.


Subject(s)
Calcification, Physiologic/physiology , Craniosynostoses/surgery , Osteogenesis/physiology , Polyesters/pharmacology , Tissue Scaffolds/chemistry , Alkaline Phosphatase/metabolism , Cell Differentiation , Coated Materials, Biocompatible/pharmacology , Humans , In Vitro Techniques , Nanotechnology , Osteoblasts/cytology , Polyethylene Glycols/pharmacology , Skull/cytology , Skull/surgery , Surface Properties , X-Ray Microtomography
2.
Orthod Craniofac Res ; 18 Suppl 1: 196-206, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25865549

ABSTRACT

OBJECTIVES: Craniosynostosis, the premature fusion of cranial bones, has traditionally been described as a disease of increased bone mineralization. However, multiple mouse models of craniosynostosis display craniosynostosis simultaneously with diminished cranial bone volume and/or density. We propose an alternative hypothesis that craniosynostosis results from abnormal tissue mineralization through the downregulation of tissue-non-specific alkaline phosphatase (TNAP) enzyme downstream of activating mutations in FGFRs. MATERIAL AND METHODS: Neonatal Crouzon (FGFRC342Y/+) and wild-type (FGFR+/+) mice were injected with lentivirus to deliver a recombinant form of TNAP. Mice were sacrificed at 4 weeks postnatal. Serum was collected to test for alkaline phosphatase (AP), phosphorus, and calcium levels. Craniofacial bone fusion and morphology were assessed by micro-computed tomography. RESULTS: Injection with the TNAP lentivirus significantly increased serum AP levels (increased serum AP levels are indicative of efficient transduction and production of the recombinant protein), but results were variable and dependent upon viral lot and the litter of mice injected. Morphological analysis revealed craniofacial form differences for inferior surface (p=0.023) and cranial height (p=0.014) regions between TNAP lentivirus-injected and vehicle-injected Crouzon mice. With each unit increase in AP level, the odds of lambdoid suture fusion decreased by 84.2% and these results came close to statistical significance (p=0.068). CONCLUSION: These results suggest that TNAP deficiency may mediate FGFR2-associated craniosynostosis. Future studies should incorporate injection of recombinant TNAP protein, to avoid potential side effects and variable efficacy of lentiviral gene delivery.


Subject(s)
Alkaline Phosphatase/genetics , Craniosynostoses/therapy , Genetic Therapy , Receptor, Fibroblast Growth Factor, Type 2/genetics , Skull/pathology , Alkaline Phosphatase/blood , Animals , Calcification, Physiologic/genetics , Calcium/blood , Cephalometry/methods , Cranial Sutures/growth & development , Cranial Sutures/pathology , Craniosynostoses/physiopathology , Cysteine/genetics , Disease Models, Animal , Female , Genetic Vectors/genetics , Lentivirus/genetics , Male , Mice , Mice, Inbred BALB C , Occipital Bone/growth & development , Occipital Bone/pathology , Parietal Bone/growth & development , Parietal Bone/pathology , Phosphorus/blood , Signal Transduction/genetics , Skull/growth & development , Tyrosine/genetics , X-Ray Microtomography/methods
3.
Orthod Craniofac Res ; 18 Suppl 1: 187-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25865548

ABSTRACT

OBJECTIVES: To determine minimal dose levels required for local inhibition of orthodontic relapse by recombinant OPG protein (OPG-Fc), while also determining effects of injected OPG-Fc on alveolar bone and long bone. SETTING AND SAMPLE POPULATION: The Department of Orthodontics and Pediatric Dentistry at the University of Michigan. Eighteen male Sprague Dawley rats. MATERIALS & METHODS: Maxillary molars were moved with nickel-titanium springs and then allowed to relapse in Sprague Dawley rats. Upon appliance removal, animals were injected with a single dose of 1.0 mg/kg OPG-Fc, 0.1 mg/kg OPG-Fc, or phosphate-buffered saline (vehicle) just distal to the molar teeth. Tooth movement measurements were made from stone casts, which were scanned and digitally measured. Alveolar tissues were examined by histology. Micro-computed tomography was used to quantify changes in alveolar and femur bone. RESULTS: Local injection of OPG-Fc inhibited molar but not incisor relapse, when compared to vehicle-injected animals. No significant differences in alveolar or femur bone were seen between the three treatment groups after 24 days of relapse. CONCLUSIONS: Our results demonstrate that a single local injection of OPG-Fc effectively inhibits orthodontic relapse, with minimal systemic bone metabolic effects. Our results also show that a single injection of OPG-Fc will influence tooth movement only in teeth close to the injection site. These findings indicate that OPG-Fc has potential as a safe and effective pharmacological means to locally control osteoclasts, for uses such as maintaining anchorage during orthodontic tooth movement and preventing orthodontic relapse in humans.


Subject(s)
Alveolar Process/drug effects , Bone Resorption/prevention & control , Osteoprotegerin/therapeutic use , Tooth Movement Techniques/methods , Alveolar Process/pathology , Animals , Bone Density/drug effects , Bone Remodeling/drug effects , Bone Resorption/pathology , Femur/drug effects , Femur/pathology , Incisor/drug effects , Injections , Male , Maxilla/drug effects , Maxilla/pathology , Models, Dental , Molar/drug effects , Orthodontic Wires , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoprotegerin/administration & dosage , Pharmaceutical Vehicles , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Recurrence , Tooth Movement Techniques/instrumentation , X-Ray Microtomography/methods
4.
J Dent Res ; 94(5): 706-14, 2015 May.
Article in English | MEDLINE | ID: mdl-25716980

ABSTRACT

Mutations in ALPL result in hypophosphatasia (HPP), a disease causing defective skeletal mineralization. ALPL encodes tissue nonspecific alkaline phosphatase (ALP), an enzyme that promotes mineralization by reducing inorganic pyrophosphate, a mineralization inhibitor. In addition to skeletal defects, HPP causes dental defects, and a mild clinical form of HPP, odontohypophosphatasia, features only a dental phenotype. The Alpl knockout (Alpl (-/-)) mouse phenocopies severe infantile HPP, including profound skeletal and dental defects. However, the severity of disease in Alpl (-/-) mice prevents analysis at advanced ages, including studies to target rescue of dental tissues. We aimed to generate a knock-in mouse model of odontohypophosphatasia with a primarily dental phenotype, based on a mutation (c.346G>A) identified in a human kindred with autosomal dominant odontohypophosphatasia. Biochemical, skeletal, and dental analyses were performed on the resulting Alpl(+/A116T) mice to validate this model. Alpl(+/A116T) mice featured 50% reduction in plasma ALP activity compared with wild-type controls. No differences in litter size, survival, or body weight were observed in Alpl(+/A116T) versus wild-type mice. The postcranial skeleton of Alpl(+/A116T) mice was normal by radiography, with no differences in femur length, cortical/trabecular structure or mineral density, or mechanical properties. Parietal bone trabecular compartment was mildly altered. Alpl(+/A116T) mice featured alterations in the alveolar bone, including radiolucencies and resorptive lesions, osteoid accumulation on the alveolar bone crest, and significant differences in several bone properties measured by micro-computed tomography. Nonsignificant changes in acellular cementum did not appear to affect periodontal attachment or function, although circulating ALP activity was correlated significantly with incisor cementum thickness. The Alpl(+/A116T) mouse is the first model of odontohypophosphatasia, providing insights on dentoalveolar development and function under reduced ALP, bringing attention to direct effects of HPP on alveolar bone, and offering a new model for testing potential dental-targeted therapies in future studies.


Subject(s)
Adenine , Gene Knock-In Techniques/methods , Hypophosphatasia/genetics , Periodontal Diseases/genetics , Thymine , Tooth Demineralization/congenital , Alkaline Phosphatase/genetics , Alveolar Process/pathology , Animals , Biomechanical Phenomena , Bone Density/physiology , Bone Matrix/pathology , Bone Resorption/pathology , Calcification, Physiologic/physiology , Dental Cementum/pathology , Disease Models, Animal , Femur/physiopathology , Humans , Male , Mandible/pathology , Mice , Parietal Bone/pathology , Pliability , Tooth Demineralization/genetics , X-Ray Microtomography/methods
5.
Biomed Res Int ; 2013: 292506, 2013.
Article in English | MEDLINE | ID: mdl-23762837

ABSTRACT

We recently reported that cranial bones of Fgfr2(C342Y/+) craniosynostotic mice are diminished in density when compared to those of wild type mice, and that cranial bone cells isolated from the mutant mice exhibit inhibited late stage osteoblast differentiation. To provide further support for the idea that craniosynostosis-associated Fgfr mutations lead to cell autonomous defects in osteoblast differentiation and mineralized tissue formation, here we tested bone marrow stromal cells isolated from Fgfr2(C342Y/+) mice for their ability to differentiate into osteoblasts. Additionally, to determine if the low bone mass phenotype of Crouzon syndrome includes the appendicular skeleton, long bones were assessed by micro CT. Fgfr2(C342Y/+) cells showed increased osteoblastic gene expression during early osteoblastic differentiation but decreased expression of alkaline phosphatase mRNA and enzyme activity, and decreased mineralization during later stages of differentiation, when cultured under 2D in vitro conditions. Cells isolated from Fgfr2(C342Y/+) mice also formed less bone when allowed to differentiate in a 3D matrix in vivo. Cortical bone parameters were diminished in long bones of Fgfr2(C342Y/+) mice. These results demonstrate that marrow stromal cells of Fgfr2(C342Y/+) mice have an autonomous defect in osteoblast differentiation and bone mineralization, and that the Fgfr2(C342Y) mutation influences both the axial and appendicular skeletons.


Subject(s)
Cell Differentiation , Craniosynostoses/genetics , Mesenchymal Stem Cells/pathology , Mutation/genetics , Osteoblasts/pathology , Osteogenesis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calcification, Physiologic , Craniosynostoses/diagnostic imaging , Craniosynostoses/pathology , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Organ Size , Osteoblasts/metabolism , Staining and Labeling , Subcutaneous Tissue/pathology , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...