Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Pharmaceutics ; 16(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38675146

ABSTRACT

Antioxidant vanillin (4-hydroxy-3-methoxybenzaldehyde) is used as a flavoring in foods, beverages, and pharmaceuticals. Vanillin possesses various biological effects, such as antioxidant, anti-inflammatory, antibacterial, and anticancer properties. This study aimed to investigate the biological activities of vanillin purified from Adenophora triphylla var. japonica Hara on bone-forming processes. Vanillin treatment induced mineralization as a marker for mature osteoblasts, after stimulating alkaline phosphatase (ALP) staining and activity. The bone-forming processes of vanillin are mainly mediated by the upregulation of the bone morphogenetic protein 2 (BMP2), phospho-Smad1/5/8, and runt-related transcription factor 2 (RUNX2) pathway during the differentiation of osteogenic cells. Moreover, vanillin promoted osteoblast-mediated bone-forming phenotypes by inducing migration and F-actin polymerization. Furthermore, we validated that vanillin-mediated bone-forming processes were attenuated by noggin and DKK1. Finally, we demonstrated that vanillin-mediated antioxidant effects prevent the death of osteoblasts during bone-forming processes. Overall, vanillin has bone-forming properties through the BMP2-mediated biological mechanism, indicating it as a bone-protective compound for bone health and bone diseases such as periodontitis and osteoporosis.

2.
Int J Mol Sci ; 22(9)2021 May 06.
Article in English | MEDLINE | ID: mdl-34066458

ABSTRACT

Paeonia suffruticosa has been extensively used as a traditional medicine with various beneficial effects; paeonolide (PALI) was isolated from its dried roots. This study aimed to investigate the novel effects and mechanisms of PALI in pre-osteoblasts. Here, cell viability was evaluated using an MTT assay. Early and late osteoblast differentiation was examined by analyzing the activity of alkaline phosphatase (ALP) and by staining it with Alizarin red S (ARS). Cell migration was assessed using wound healing and Boyden chamber assays. Western blot and immunofluorescence analyses were used to examine the intracellular signaling pathways and differentiation proteins. PALI (0.1, 1, 10, 30, and 100 µM) showed no cytotoxic or proliferative effects in pre-osteoblasts. In the absence of cytotoxicity, PALI (1, 10, and 30 µM) promoted wound healing and transmigration during osteoblast differentiation. ALP staining demonstrated that PALI (1, 10, and 30 µM) promoted early osteoblast differentiation in a dose-dependent manner, and ARS staining showed an enhanced mineralized nodule formation, a key indicator of late osteoblast differentiation. Additionally, low concentrations of PALI (1 and 10 µM) increased the bone morphogenetic protein (BMP)-Smad1/5/8 and Wnt-ß-catenin pathways in osteoblast differentiation. Particularly, PALI (1 and 10 µM) increased the phosphorylation of ERK1/2 compared with BMP2 treatment, an FDA-approved drug for bone diseases. Furthermore, PALI-mediated early and late osteoblast differentiation was abolished in the presence of the ERK1/2 inhibitor U0126. PALI-induced RUNX2 (Cbfa1) expression and nuclear localization were also attenuated by blocking the ERK1/2 pathway during osteoblast differentiation. We suggest that PALI has biologically novel activities, such as enhanced osteoblast differentiation and bone mineralization mainly through the intracellular ERK1/2-RUNX2 signaling pathway, suggesting that PALI might have therapeutic action and aid the treatment and prevention of bone diseases, such as osteoporosis and periodontitis.


Subject(s)
Acetophenones/pharmacology , Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/metabolism , Osteogenesis , Animals , Bone Morphogenetic Protein 2/metabolism , Calcification, Physiologic/drug effects , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , MAP Kinase Signaling System/drug effects , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Wnt3 Protein/metabolism
3.
Exp Cell Res ; 399(2): 112456, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33417921

ABSTRACT

Identifying patient mutations driving skeletal development disorders has driven our understanding of bone development. Integrin adhesion deficiency disease is caused by a Kindlin-3 (fermitin family member 3) mutation, and its inactivation results in bleeding disorders and osteopenia. In this study, we uncover a role for Kindlin-3 in the differentiation of bone marrow mesenchymal stem cells (BMSCs) down the chondrogenic lineage. Kindlin-3 expression increased with chondrogenic differentiation, similar to RUNX2. BMSCs isolated from a Kindlin-3 deficient patient expressed chondrocyte markers, including SOX9, under basal conditions, which were further enhanced with chondrogenic differentiation. Rescue of integrin activation by a constitutively activated ß3 integrin construct increased adhesion to multiple extracellular matrices and reduced SOX9 expression to basal levels. Growth plates from mice expressing a mutated Kindlin-3 with the integrin binding site ablated demonstrated alterations in chondrocyte maturation similar to that seen with the human Kindlin-3 deficient BMSCs. These findings suggest that Kindlin-3 expression mirrors RUNX2 during chondrogenesis.


Subject(s)
Chondrogenesis/genetics , Cytoskeletal Proteins/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/physiology , Neoplasm Proteins/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Mutation/physiology
4.
Arch Med Sci ; 16(6): 1419-1431, 2020.
Article in English | MEDLINE | ID: mdl-33224342

ABSTRACT

INTRODUCTION: In senile osteoporosis countering the age-mediated bone loss, promotion of osteoblastogenesis and identification of responsible micro-RNA (miR) would be a successful strategy. MATERIAL AND METHODS: miR microarray screening was carried out to identify the suppressed miRs after real time polymerase chain reaction (RT-PCR) analysis in mesenchymal stem cells (MSCs) derived from adult bone marrow during the proliferation to the mineralization stage. The primary calvarial pre-osteoblasts (human) were harvested and received transfection of miR-22's antagomir or agomir in vitro. Bioinformatics study suggested YWHAZ as the favorable target gene. Next, YWHAZ knockdown was studied for its effect on differentiation of osteoblasts. For in vivo studies, ovariectomized or sham mice were injected with miR-22's antagomir for a period of 6 weeks. The stromal cells were isolated in the 6th week for ex vivo experiments. RESULTS: miR-22 was found to be down-regulated in bone marrow derived mesenchymal stem cells. miR-22's antagomir converted the pre-osteoblasts to a more differentiated and mineralized phenotype showing upregulated protein expression of COL1A1, ALP and CBFA1. The miR-22's antagomir suppressed YWHAZ, enhanced stability of CBFA1 and promoted the differentiation of osteoblasts. In vivo, miR-22's antagomir promoted mineralization and osteoblastogenesis, elevated bone strength and reversed the ovariectomy mediated bone loss in sham mice. CONCLUSIONS: Inhibition of miR-22 may be a potential target for treating osteoporosis clinically. The findings hence suggest that inhibition of miR-22 may be an effective anabolic therapeutic approach in treating osteoporosis clinically.

5.
Rev Med Liege ; 75(10): 639-643, 2020 Oct.
Article in French | MEDLINE | ID: mdl-33030838

ABSTRACT

The Pierre-Marie Sainton syndrome or cleidocranial dysplasia is a rare congenital malformation due to a mutation in the RUNX2 gene, causing disruption in osteoblastic maturation, which results in various skeletal, dental and endocrine abnormalities. These various disorders may also have otorhinolaryngology and psychological consequences. We report the case of a patient with this rare birth defect.


La maladie de Marie et Sainton, ou dysostose cleido-crânienne, est une malformation congénitale héré-ditaire rare liée à une mutation du gène RUNX2, entraînant un déficit de maturation ostéoblastique avec, pour conséquences, différentes anomalies squelettiques, dentaires et endocrinologiques. Ces différents troubles peuvent également avoir des répercussions sur le plan ORL et psychologique. Nous rapportons le cas d'un patient présentant cette malformation congénitale rare.


Subject(s)
Cleidocranial Dysplasia , Cleidocranial Dysplasia/diagnosis , Cleidocranial Dysplasia/genetics , Humans , Mutation
6.
Rev. ADM ; 77(4): 222-226, jul.-ago. 2020. ilus
Article in Spanish | LILACS | ID: biblio-1130183

ABSTRACT

Introducción: La disostosis cleidocraneal es un trastorno genético raro con patrón hereditario. Sus rasgos patognomónicos son la aplasia clavicular, fontanelas y suturas abiertas, múltiples anomalías dentales. Su origen se relaciona con alteraciones en el gen RUNX2, importante para la síntesis de CBFA1, que a su vez funciona como un conformador óseo y un diferenciador de osteoblastos. Caso clínico: Paciente de 11 años con características clínicas de CCD, se corroboran los antecedentes genéticos hereditarios y alteraciones dentales relacionados con disostosis cleidocraneal. Resultado: A tres años de tratamiento se observa mejor definición facial, la erupción de piezas permanentes retenidas y mejor función masticatoria. Conclusión: Mejorar la calidad de vida del paciente con tratamientos interceptivos y el conocimiento de las alteraciones causadas por el síndrome, así como el trabajo interdisciplinario (AU)


Introduction: Cleidocranial dysostosis is a rare genetic disorder with a hereditary pattern. Its pathognomonic features are clavicular aplasia, fontanelles and open sutures, multiple dental anomalies. Its origin is related to alterations in the RUNX2 gene, important for the synthesis of CBFA1, which in turn functions as a bone conformer and an osteoblast differentiator. Clinical case: Patient with eleven years old with clinical characteristics of CCD, hereditary genetic background, and dental alterations related to cleidocranial dysostosis are corroborated. Result: After three years of treatment, the facial definition is better, the eruption of permanent pieces retained and better chewing function. Conclusion: Improve the quality of life of the patient with interceptive treatments and the knowledge of the alterations caused by the syndrome, as well as the interdisciplinary work (AU)


Subject(s)
Humans , Female , Child , Orthodontics, Interceptive , Tooth Abnormalities/therapy , Cleidocranial Dysplasia/therapy , Genetic Diseases, Inborn , Patient Care Team , Quality of Life , Schools, Dental , Tooth Eruption , Mexico
7.
BMC Mol Cell Biol ; 21(1): 27, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32295522

ABSTRACT

BACKGROUND: Changes in gene regulation are widely recognized as an important driver of adaptive phenotypic evolution. However, the specific molecular mechanisms that underpin such changes are still poorly understood. Chromatin state plays an essential role in gene regulation, by influencing the accessibility of coding loci to the transcriptional machinery. Changes in the function of chromatin remodellers are therefore strong candidates to drive changes in gene expression associated with phenotypic adaptation. Here, we identify amino acid homoplasies in the chromatin remodeller CHD9, shared between the extinct marsupial thylacine and eutherian wolf which show remarkable skull convergence. CHD9 is involved in osteogenesis, though its role in the process is still poorly understood. We examine whether CHD9 is able to regulate the expression of osteogenic target genes and examine the function of a key substitution in the CHD9 DNA binding domain. RESULTS: We examined whether CHD9 was able to upregulate its osteogenic target genes, RUNX2, Osteocalcin (OC) and ALP in HEK293T cells. We found that overexpression of CHD9 upregulated RUNX2, the master regulator of osteoblast cell fate, but not the downstream genes OC or ALP, supporting the idea that CHD9 regulates osteogenic progenitors rather than terminal osteoblasts. We also found that the evolutionary substitution in the CHD9 DNA binding domain does not alter protein secondary structure, but was able to drive a small but insignificant increase in RUNX2 activation. Finally, CHD9 was unable to activate an episomal RUNX2 promoter-reporter construct, suggesting that CHD9 requires the full chromatin complement for its function. CONCLUSIONS: We provide new evidence to the role of CHD9 in osteogenic differentiation through its newly observed ability to upregulate the expression of RUNX2. Though we were unable to identify significant functional consequences of the evolutionary substitution in HEK293T cells, our study provides important steps forward in the functional investigation of protein homoplasy and its role in developmental processes. Mutations in coding genes may be a mechanism for driving adaptive changes in gene expression, and their validation is essential towards determining the functional consequences of evolutionary homoplasy.


Subject(s)
Core Binding Factor Alpha 1 Subunit , DNA Helicases/metabolism , Trans-Activators/metabolism , Biological Evolution , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Evolution, Molecular , Gene Expression Regulation , HEK293 Cells , Humans , Osteogenesis/genetics , Skeleton , Transcriptional Activation
8.
Clin Genet ; 96(5): 429-438, 2019 11.
Article in English | MEDLINE | ID: mdl-31347140

ABSTRACT

RUNX2 (Runt-related transcription factor 2) is a master regulator of osteoblast differentiation, cartilage and bone development. Pathogenic variants in RUNX2 have been linked to the Cleidocranial dysplasia (CCD), which is characterized by hypoplasia or aplasia of clavicles, delayed fontanelle closure, and dental anomalies. Here, we report 11 unrelated Polish patients with CCD caused by pathogenic alterations located in the Runt domain of RUNX2. In total, we identified eight different intragenic variants, including seven missense and one splicing mutation. Three of them are novel: c.407T>A p.(Leu136Gln), c.480C>G p.(Asn160Lys), c.659C>G p.(Thr220Arg), additional three were not functionally tested: c.391C>T p.(Arg131Cys), c.580+1G>T p.(Lys195_Arg229del), c.652A>G p.(Lys218Glu), and the remaining two: c.568C>T p.(Arg190Trp), c.673C>T p.(Arg225Trp) were previously reported and characterized. The performed transactivation and localization studies provide evidence of decreased transcriptional activity of RUNX2 due to mutations targeting the Runt domain and prove that impairment of nuclear localization signal (NLS) affects the subcellular localization of the protein. Presented data show that pathogenic variants discovered in our patients have a detrimental effect on RUNX2, triggering the CCD phenotype.


Subject(s)
Cleidocranial Dysplasia/genetics , Core Binding Factor Alpha 1 Subunit/chemistry , Genetic Predisposition to Disease , Protein Conformation , Child, Preschool , Cleidocranial Dysplasia/epidemiology , Cleidocranial Dysplasia/pathology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/ultrastructure , Female , Humans , Infant , Male , Mutation/genetics , Mutation, Missense/genetics , Phenotype , Poland/epidemiology , Protein Isoforms/genetics , Structure-Activity Relationship
9.
BMC Evol Biol ; 17(1): 110, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28472940

ABSTRACT

BACKGROUND: Runt-related transcription factor 2 (RUNX2) is a transcription factor essential for skeletal development. Variation within the RUNX2 polyglutamine / polyalanine (QA) repeat is correlated with facial length within orders of placental mammals and is suggested to be a major driver of craniofacial diversity. However, it is not known if this correlation exists outside of the placental mammals. RESULTS: Here we examined the correlation between the RUNX2 QA repeat ratio and facial length in the naturally evolving sister group to the placental mammals, the marsupials. Marsupials have a diverse range of facial lengths similar to that seen in placental mammals. Despite their diversity there was almost no variation seen in the RUNX2 QA repeat across individuals spanning the entire marsupial infraclass. The extreme conservation of the marsupial RUNX2 QA repeat indicates it is under strong purifying selection. Despite this, we observed an unexpectedly high level of repeat purity. CONCLUSIONS: Unlike within orders of placental mammals, RUNX2 repeat variation cannot drive craniofacial diversity in marsupials. We propose conservation of the marsupial RUNX2 QA repeat is driven by the constraint of accelerated ossification of the anterior skeleton to facilitate life in the pouch. Thus, marsupials must utilize alternate pathways to placental mammals to drive craniofacial evolution.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Marsupialia/anatomy & histology , Marsupialia/genetics , Skull/anatomy & histology , Animals , Biological Evolution , Mammals/anatomy & histology , Mammals/classification , Mammals/genetics , Marsupialia/classification , Osteogenesis , Repetitive Sequences, Nucleic Acid
10.
Clin Genet ; 90(5): 393-402, 2016 11.
Article in English | MEDLINE | ID: mdl-27272193

ABSTRACT

Runt-related transcription factor 2 (RUNX2/Cbfa1) is the main regulatory gene controlling skeletal development and morphogenesis in vertebrates. It is located on chromosome 6p21 and has two functional isoforms (type I and type II) under control of two alternate promoters (P1 and P2). Mutations within RUNX2 are linked to Cleidocranial dysplasia syndrome (CCD) in humans. CCD is an autosomal skeletal disorder characterized by several features such as delayed closure of fontanels, dental abnormalities and hypoplastic clavicles. Here, we summarize recent knowledge about RUNX2 function, mutations and their phenotypic consequences in patients.


Subject(s)
Cleidocranial Dysplasia/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Mutation , Cleidocranial Dysplasia/physiopathology , Genetic Association Studies , Heterozygote , Humans , Pedigree , Phenotype
11.
J Orthop Case Rep ; 5(2): 21-4, 2015.
Article in English | MEDLINE | ID: mdl-27299035

ABSTRACT

INTRODUCTION: Cleidocranial dysplasia (CCD) is characterized by aplasia or hypoplasia of the clavicles, characteristic craniofacial malformations, and the presence of numerous supernumerary and unerupted teeth. It affects bones derived from both intra-membranous and endochondral ossification. Incidence has been reported as 1 in 10,00,000. It is caused by mutation in the gene encoding transcription factor Core Binding Factor Subunit Alpha l (CBFAl) or Runt related transcription factor 2 (RUNX2). CASE REPORT: This presentation discusses the clinical and radiographic features of a familial case of cleidocranial dysplasia occurring in a father and a child. All the clinical and radiographic features, except that of the chest x-ray, were more prominent in the child than the father. This supports the fact that CCD is transmitted by an autosomal-dominant mode of inheritance with high penetrance and variable expressivity. It is sporadic in about 40% of cases. Each child of an individual with CCD has a 50% chance of in heriting the mutation. CONCLUSION: Diagnosis is mostly made on the basis of clinical and radiographic features. Molecular genetic testing such as sequence analysis or deletion analysis can be used in cleidocranial dysplasia. Some cases are diagnosed through incidental findings by physicians, treating patients for unrelated conditions. Treatment of these patients requires a multidisciplinary approach which includes orthopaedic and dental corrections along with management of any complications of cleidocranial dysplasia.

12.
Biomatter ; 4: e967616, 2014.
Article in English | MEDLINE | ID: mdl-25482335

ABSTRACT

Magnesium-based implants exhibit various advantages such as biodegradability and potential for enhanced in vivo bone formation. However, the cellular mechanisms behind this possible osteoconductivity remain unclear. To determine whether high local magnesium concentrations can be osteoconductive and exclude other environmental factors that occur during the degradation of magnesium implants, magnesium salt (MgCl2) was used as a model system. Because cell lines are preferred targets in studies of non-degradable implant materials, we performed a comparative study of 3 osteosarcoma-derived cell lines (MG63, SaoS2 and U2OS) with primary human osteoblasts. The correlation among cell count, viability, cell size and several MgCl2 concentrations was used to examine the influence of magnesium on proliferation in vitro. Moreover, bone metabolism alterations during proliferation were investigated by analyzing the expression of genes involved in osteogenesis. It was observed that for all cell types, the cell count decreases at concentrations above 10 mM MgCl2. However, detailed analysis showed that MgCl2 has a relevant but very diverse influence on proliferation and bone metabolism, depending on the cell type. Only for primary cells was a clear stimulating effect observed. Therefore, reliable results demonstrating the osteoconductivity of magnesium implants can only be achieved with primary osteoblasts.


Subject(s)
Magnesium Chloride/pharmacology , Osteoblasts/drug effects , Osteogenesis/drug effects , Cell Count , Cell Line , Cell Proliferation , Cell Size , Cell Survival , Gene Expression Regulation/drug effects , Humans , Osmolar Concentration , Osteoblasts/pathology , Osteoblasts/physiology
13.
Organogenesis ; 10(3): 289-98, 2014.
Article in English | MEDLINE | ID: mdl-25482504

ABSTRACT

A recent paper demonstrated that decellularized extracellular matrix (DECM) deposited by synovium-derived stem cells (SDSCs), especially from fetal donors, could rejuvenate human adult SDSCs in both proliferation and chondrogenic potential, in which expanded cells and corresponding culture substrate (such as DECM) were found to share a mutual reaction in both elasticity and protein profiles (see ref. (1) ). It seems that young DECM may assist in the development of culture strategies that optimize proliferation and maintain "stemness" of mesenchymal stem cells (MSCs), helping to overcome one of the primary difficulties in MSC-based regenerative therapies. In this paper, the effects of age on the proliferative capacity and differentiation potential of MSCs are reviewed, along with the ability of DECM from young cells to rejuvenate old cells. In an effort to highlight some of the potential molecular mechanisms responsible for this phenomenon, we discuss age-related changes to extracellular matrix (ECM)'s physical properties and chemical composition.


Subject(s)
Cell Communication/physiology , Cellular Senescence/physiology , Extracellular Matrix/physiology , Mesenchymal Stem Cells , Tissue Engineering , Animals , Cell Proliferation , Cells, Cultured , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Rats , Stem Cell Niche
14.
Bone ; 69: 126-32, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25263523

ABSTRACT

Mechanical stimulation and biological factors coordinately regulate bone development and regeneration; however, the underlying mechanisms are poorly understood. Microgravity induces bone loss, which may be partly related to the development of resistance to local cytokines, including insulin-like growth factor 1 (IGF-1). Here, we report the involvement of integrin αvß3 in microgravity-associated bone loss. An established OSE-3T3 cell model was stably transfected with a 6OSE2 (Osteoblast-Specific Element 2)-luciferase reporter and cultured under simulated microgravity (SMG) and hypergravity (HG) conditions in the presence or absence of IGF-1, the disintegrin echistatin, the phosphoinositide 3-kinase (PI3K) inhibitor LY294002, or combinations of these agents. Activity of core-binding factor α1 (Cbfa1), an essential transcription factor for osteoblastic differentiation and osteogenesis, was reflected by luciferase activity. Different gravity conditions affected the induction of IGF-1 and subsequent effects on Cbfa1 transcription activity. SMG and HG influenced the expression and activity of integrin αvß3 and phosphorylation level of p85. LY294002 inhibited the effects of HG or IGF-1 on Cbfa1 activity, indicating that HG and IGF-1 could increase Cbfa1 activity via PI3K signaling. Inhibition of integrin αvß3 by echistatin attenuated the induction of IGF-1 and thus its effect on Cbfa1 activity under normal and HG conditions. Co-immunoprecipitation demonstrated that integrin ß3 interacted with insulin receptor substrate 1, and that this interaction was decreased under SMG and increased under HG conditions. These results suggest that integrin αvß3 mediates the synergetic regulation of Cbfa1 transcription activity by gravity and IGF-1 via PI3K signaling.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Gravity, Altered/adverse effects , Insulin-Like Growth Factor I/metabolism , Integrin alphaVbeta3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , 3T3 Cells , Animals , Blotting, Western , Bone Remodeling/physiology , Immunoprecipitation , Mice , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Space Flight , Transfection , Weightlessness/adverse effects
15.
Biomaterials ; 35(28): 8236-48, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24965885

ABSTRACT

During stem cell differentiation, various cellular responses occur that are mediated by transcription factors and proteins. This study evaluated the abilities of SOX9, a crucial protein during the early stage of chondrogenesis, and siRNA targeting Cbfa-1, a transcription factor that promotes osteogenesis, to stimulate chondrogenesis. Non-toxic poly-(d,l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) were coated with Cbfa-1-targeting siRNA and loaded with SOX9 protein. Coomassie blue staining and circular dichroism revealed that the loaded SOX9 protein maintained its stability and bioactivity. These NPs easily entered human mesenchymal stem cells (hMSCs) in vitro and caused them to differentiate into chondrocytes. Markers that are typically expressed in mature chondrocytes were examined. These markers were highly expressed at the mRNA and protein levels in hMSCs treated with PLGA NPs coated with Cbfa-1-targeting siRNA and loaded with SOX9 protein. By contrast, these cells did not express osteogenesis-related markers. hMSCs were injected into mice following internalization of PLGA NPs coated with Cbfa-1-targeting siRNA and loaded with SOX9 protein. When the injection site was excised, markers of chondrogenesis were found to be highly expressed at the mRNA and protein levels, similar to the in vitro results. When hMSCs internalized these NPs and were then cultured in vitro or injected into mice, chondrogenesis-related extracellular matrix components were highly expressed.


Subject(s)
Chondrogenesis/drug effects , Core Binding Factor Alpha 1 Subunit/chemistry , Lactic Acid/chemistry , Mesenchymal Stem Cells/cytology , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , RNA, Small Interfering/chemistry , SOX9 Transcription Factor/chemistry , Animals , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Circular Dichroism , Drug Delivery Systems , Extracellular Matrix/metabolism , Female , Fluorescein-5-isothiocyanate/chemistry , Gene Transfer Techniques , Glycosaminoglycans/chemistry , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Microscopy, Electron, Scanning , Nanotechnology/methods , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Messenger/metabolism
16.
J Trace Elem Med Biol ; 28(3): 278-83, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24680482

ABSTRACT

To study the effects and importance of fluoride on FBs in the development of extraperiosteal calcification and the ossification of skeletal fluorosis, the presence of the osteogenic phenotype, which is indicated by the expression of core-binding factor α1 (Cbfa1) and osteocalcin (OCN), in an FB cell line (L929) and in osteoblasts (OBs) exposed to fluoride was determined. Fibroblasts and osteoblasts were exposed to different concentrations of fluoride (0, 0.0001, 0.001, 0.1, 1.0, 10.0 and 20.0 mg/L F(-)). By using RT-PCR and ELISA, the mRNA levels of Cbfa1 and OCN were measured at 48 h, and the protein levels of Cbfa1 and OCN were measured at 2, 4, 24, 48 and 72 h. The data demonstrated the following: (1) The Cbfa1 protein level in fluoride-treated fibroblasts clearly increased at 48 h in the groups treated with 0.0001, 0.001, 0.1, 1.0 and 20.0 mg/L F(-). The Cbfa1 protein level of the group treated with 10 mg/L F(-) at 72 h was higher than that of the control group. The level of Cbfa1 mRNA in the fibroblasts was much higher at 48 h in the group treated with 10.0 mg/L F(-) than in the control group. (2) The OCN protein level in fluoride-treated fibroblasts was significantly higher than that of the control group in the 0.0001, 0.1, 1.0, 10.0 and 20.0 mg/L F(-) groups at 2 h, and in the 0.001 and 0.1 F(-) groups at 4 h. A slightly higher level of OCN mRNA in fluoride-treated fibroblasts was also found in the 1.0 and 20.0 mg/L F(-) groups compared to the control group. (3) The expressions of Cbfa1 and OCN in osteoblasts treated with the same experimental conditions as the fibroblasts were up-regulated by fluoride following the same trend as in the fibroblasts. Our results showed an increase in the expression of Cbfa1 and OCN in fibroblasts and osteoblasts exposed to fluoride and suggested that the osteogenic function of fibroblasts induced by fluoride could play an important role in the development of extraperiosteal ossification during skeletal fluorosis.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Fibroblasts/metabolism , Osteoblasts/metabolism , Osteocalcin/metabolism , Animals , Cell Line , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fibroblasts/drug effects , Fluorides/pharmacology , Immunohistochemistry , Mice , Osteoblasts/drug effects
17.
Chem Biol Interact ; 207: 26-31, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-24239970

ABSTRACT

Glucocorticoids therapy is strongly limited since extended glucocorticoids can cause serious side effects, including increased susceptibility to develop the bone disease osteoporosis. Despite its side effects recognized importance to clinicians, seldom is known about how glucocorticoids directly impact bone-forming osteoblasts. Previous studies showed that dexamethasone (DEX) induces excessive production of reactive oxygen species (ROS), and causes oxidative stress in rat hippocampal slice cultures. To assess the implications and investigate the mechanisms of glucocorticoid-elicited osteoporosis, we hypothesize that DEX exposure induces oxidative stress which leads to decreased Cbfa1 mRNA expression, and predict that the antioxidant N-acetylcysteine (NAC) mitigates the damaging effects of DEX. Oxidative stress is implicated in osteoporosis. Furthermore, the osteoblast transcriptional factor Cbfa1 is reported to play a protective role against osteoporosis in postmenopausal women. Cells treated with (0.1, 1, 10µM) DEX exhibited signs of oxidative damages including depletion in total antioxidant capacity (T-AOC), increased ROS formation, and enhanced lipid peroxidation. Cbfa1 mRNA expression, by RT-PCR, was significantly reduced after exposure to (0.1, 1, 10µM) DEX. Pretreatment with the antioxidant NAC (2mM) prevented DEX-induced decrease in Cbfa1 mRNA. This study provides insight into the underlying mechanisms of high dose DEX-induced osteotoxicity. DEX (0.1, 1, 10µM) decreases the expression of Cbfa1 mRNA and inhibits differentiation and function of osteoblasts by inducing oxidative stress. The antioxidant NAC can mitigate the oxidative stress damaging effects of DEX. In addition, this study distinguishes itself by identifying Cbfa1 as a target for high dose DEX-induced osteotoxicity.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Glucocorticoids/pharmacology , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Alkaline Phosphatase/metabolism , Animals , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Cell Proliferation/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteocalcin/metabolism , Oxidative Stress/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Staining and Labeling
18.
Adv Biomed Res ; 1: 8, 2012.
Article in English | MEDLINE | ID: mdl-23210067

ABSTRACT

BACKGROUND: There are significant limitations in repair of irrecoverable bone defects. Stem-cell therapy is a promising approach for the construction of bone tissue. Mesenchymal stem cells (MSCs) have been introduced as basic tools for bone tissue generation. Through MSCs, adipose-derived stem cells (ADSCs) are more interesting. Since the similarity of native osteoblasts and differentiated osteoblasts from ADSCs in terms of gene expression pattern is unknown, this study was designed to compare gene expression patterns of some genes involved in osteogenesis between human native osteoblasts and adipose-derived differentiated osteoblasts. MATERIALS AND METHODS: Realtime qRT-PCR was used for studying the gene expression of osteocalcin, osteopontin, and core binding factor alpha 1 (Cbfa1) in human native osteoblasts and adipose derived osteogenic osteoblasts at days 7, 14, 21, and 28 of differentiation. RESULTS: This study demonstrated that native osteoblasts and differentiated osteoblasts, cultured in common osteogenic medium, have significant differences in gene expression levels for osteocalcin and osteopontin. Compared to native osteoblasts, these genes are expressed lower in all four groups of differentiated osteoblastic cells. We also found, there is a progressive increase in cbfa1 expression over the differentiation period of ADSCs from day 7 to day 28. CONCLUSIONS: Our findings help for better assessment of adipose-derived differentiated cells as a source for cell-based therapy.

19.
J Indian Prosthodont Soc ; 12(4): 252-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-24293923

ABSTRACT

Cleidocranial dysostosis (CCD) is an uncommon, generalized skeletal disorder characterized by delayed ossification of the skull, aplastic or hypoplastic clavicles, and complex dental abnormalities such as retention of multiple deciduous teeth, impaction or delayed eruption of permanent teeth and presence of supernumerary teeth. This case report describes a 30-year old male patient of CCD with classical findings and the challenges faced in his prosthodontic rehabilitation. Conventional complete dentures following interim complete dentures therapy had to be given as all other treatment modalities were ruled out because of anatomic limitations. Despite the disadvantages of removable dentures the patient adapted admirably to them with significant improvement in self-esteem. Though orthodontic and surgical correction may be the treatment of choice, the need to preserve the alveolar ridge could make fabrication of removable dentures in such patients, a viable option.

20.
Int J Mol Sci ; 12(7): 4206-13, 2011.
Article in English | MEDLINE | ID: mdl-21845073

ABSTRACT

Hypertension is a risk factor for osteoporosis. Animal and epidemiological studies demonstrate that high blood pressure is associated with increased calcium loss, elevated parathyroid hormone, and increased calcium movement from bone. However, the mechanism responsible for hypertension-related osteoporosis remains elusive. Recent epidemiological studies indicate the benefits of Angiotensin II Receptors Blockade (ARB) on decreasing fracture risks. Since receptors for angiotensin II, the targets of ARB, are expressed in both osteoblasts and osteoclasts, we postulated that angiotensin II plays an important role in hypertension-related osteoporosis. Cbfa1 and RANKL, the important factors for maintaining bone homeostasis and key mediators in controlling osteoblast and osteoclast differentiation, are both regulated by cAMP-dependent signaling. Angiotensin II along with factors such as LDL, HDL, NO and homocysteine that are commonly altered both in hypertension and osteoporosis, can down-regulate the expression of Cbfa1 but up-regulate RANKL expression via the cAMP signaling pathway. We thus hypothesized that, by altering the ratio of Cbfa1/RANKL expression via the cAMP-dependent pathway, angiotensin II differently regulates osteoblast and osteoclast differentiation leading to enhanced bone resorption and reduced bone formation. Since ARB can antagonize the adverse effect of angiotensin II on bone by lowering cAMP levels and modifying other downstream targets, including LDL, HDL, NO and Cbfa1/RANKL, we propose the hypothesis that the antagonistic effects of ARB may also be exerted via cAMP signaling pathway.


Subject(s)
Angiotensin II Type 2 Receptor Blockers/pharmacology , Angiotensin II/pharmacology , Core Binding Factor Alpha 1 Subunit/metabolism , Cyclic AMP/metabolism , RANK Ligand/metabolism , Signal Transduction/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hypertension/complications , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Nitric Oxide/blood , Osteoporosis/etiology , Receptor, Angiotensin, Type 2/chemistry , Receptor, Angiotensin, Type 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...