Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Circ J ; 81(7): 1043-1050, 2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28344201

ABSTRACT

BACKGROUND: Valve calcification is well estimated by ex-vivo micro-computed tomography (micro-CT). The objective of this study was to investigate the associations between micro-CT findings and biological indices of calcification in aortic stenosis (AS), as well as differences between bicuspid aortic valve (BAV) and tricuspid aortic valve (TAV).Methods and Results:Aortic valves and plasma were obtained from patients undergoing valve surgery. Valves were dissected and underwent micro-CT, genetic analyses, and calcium content assessment. Plasma levels of calcification markers were measured. Forty-two patients with isolated severe AS, including 22 with BAV, were studied. BAV patients had a lower median CT value (140.0 [130.0-152.0] vs. 157.0 [147.0-176.0], P=0.002) and high-density calcification (HDC) fraction (9.3 [5.7-23.3] % vs. 21.3 [14.3-31.2] %, P=0.01), as compared with TAV. Calcification fraction (CF) correlated with AS severity (measured as maximal transvalvular pressure gradient [r=0.34, P=0.03], maximal flow velocity [r=0.38, P=0.02], and indexed aortic valve area [r=-0.37, P=0.02]). For TAV patients only, mRNA expression of integrin-binding sialoprotein correlated with CF (r=0.45, P=0.048), and the receptor activator of the nuclear factor κ-B ligand transcript correlated with HDC corrugation (r=0.54, P=0.01). CONCLUSIONS: TAV patients with AS present more mineralized calcifications in micro-CT than BAV subjects. The relative volume of calcifications increases with the AS severity. In TAV patients, upregulated expression of genes involved in osteoblastogenesis in AS correlates with leaflet mineralization in micro-CT.


Subject(s)
Aortic Valve Stenosis , Integrin-Binding Sialoprotein/biosynthesis , Mitral Valve , RANK Ligand/biosynthesis , Tricuspid Valve , Vascular Calcification , X-Ray Microtomography , Aged , Aortic Valve Stenosis/diagnostic imaging , Aortic Valve Stenosis/metabolism , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Mitral Valve/diagnostic imaging , Mitral Valve/metabolism , Tricuspid Valve/diagnostic imaging , Tricuspid Valve/metabolism , Vascular Calcification/diagnostic imaging , Vascular Calcification/metabolism
2.
J Cell Physiol ; 230(3): 568-77, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25160656

ABSTRACT

Osteopontin (OPN) and bone sialoprotein (BSP) are coexpressed in osteoblasts and osteoclasts, and display overlapping properties. We used daily injection of parathyroid hormone 1-84 (iPTH) over the calvaria of BSP knockout (-/-) mice to investigate further their functional specificity and redundancy. iPTH stimulated bone formation in both +/+ and -/- mice, increasing to the same degree periosteum, osteoid and total bone thickness. Expression of OPN, osterix, osteocalcin (OCN) and DMP1 was also increased by iPTH in both genotypes. In contrast to +/+, calvaria cell cultures from -/- mice revealed few osteoblast colonies, no mineralization and little expression of OCN, MEPE or DMP1. In contrast, OPN levels were 5× higher in -/- versus +/+ cultures. iPTH increased alkaline phosphatase (ALP) activity in cell cultures of both genotypes, with higher OCN and the induction of mineralization in -/- cultures. siRNA blocking of OPN expression did not alter the anabolic action of the hormone in BSP +/+ calvaria, while it blunted iPTH effects in -/- mice, reduced to a modest increase in periosteum thickness. In -/- (not +/+) cell cultures, siOPN blocked the stimulation by iPTH of ALP activity and OCN expression, as well as the induction of mineralization. Thus, full expression of either OPN or BSP is necessary for the anabolic effect of PTH at least in the ectopic calvaria injection model. This suggests that OPN may compensate for the lack of BSP in the response to this hormonal challenge, and provides evidence of functional overlap between these cognate proteins.


Subject(s)
Integrin-Binding Sialoprotein/genetics , Osteogenesis/genetics , Osteopontin/genetics , Skull/growth & development , Animals , Cells, Cultured , Gene Expression Regulation, Developmental/drug effects , Integrin-Binding Sialoprotein/antagonists & inhibitors , Integrin-Binding Sialoprotein/biosynthesis , Mice , Osteogenesis/drug effects , Osteopontin/antagonists & inhibitors , Osteopontin/biosynthesis , Parathyroid Hormone/administration & dosage , RNA, Messenger/metabolism , Skull/drug effects
3.
Development ; 139(3): 601-11, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22190639

ABSTRACT

Atf4 is a leucine zipper-containing transcription factor that activates osteocalcin (Ocn) in osteoblasts and indian hedgehog (Ihh) in chondrocytes. The relative contribution of Atf4 in chondrocytes and osteoblasts to the regulation of skeletal development and bone formation is poorly understood. Investigations of the Atf4(-/-);Col2a1-Atf4 mouse model, in which Atf4 is selectively overexpressed in chondrocytes in an Atf4-null background, demonstrate that chondrocyte-derived Atf4 regulates osteogenesis during development and bone remodeling postnatally. Atf4 overexpression in chondrocytes of the Atf4(-/-);Col2a1-Atf4 double mutants corrects the reduction in stature and limb in Atf4(-/-) embryos and rectifies the decrease in Ihh expression, Hh signaling, proliferation and accelerated hypertrophy that characterize the Atf4(-/-) developing growth plate cartilages. Unexpectedly, this genetic manipulation also restores the expression of osteoblastic marker genes, namely Ocn and bone sialoprotein, in Atf4(-/-) developing bones. In Atf4(-/-);Col2a1-Atf4 adult mice, all the defective bone parameters found in Atf4(-/-) mice, including bone volume, trabecular number and thickness, and bone formation rate, are rescued. In addition, the conditioned media of ex vivo cultures from wild-type or Atf4(-/-);Col2a1-Atf4, but not Atf4(-/-) cartilage, corrects the differentiation defects of Atf4(-/-) bone marrow stromal cells and Ihh-blocking antibody eliminates this effect. Together, these data indicate that Atf4 in chondrocytes is required for normal Ihh expression and for its paracrine effect on osteoblast differentiation. Therefore, the cell-autonomous role of Atf4 in chondrocytes dominates the role of Atf4 in osteoblasts during development for the control of early osteogenesis and skeletal growth.


Subject(s)
Activating Transcription Factor 4/metabolism , Cell Differentiation , Chondrocytes/metabolism , Hedgehog Proteins/metabolism , Osteoblasts/cytology , Activating Transcription Factor 4/genetics , Animals , Bone Development , Cells, Cultured , Collagen Type II/genetics , Collagen Type II/metabolism , Female , Growth Plate/metabolism , Integrin-Binding Sialoprotein/biosynthesis , Male , Mice , Mice, Transgenic , Osteoblasts/metabolism , Osteocalcin/biosynthesis , Osteogenesis
4.
J Clin Periodontol ; 40(1): 73-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23137193

ABSTRACT

AIMS: To characterize the mineralized tissue formed constitutively in the supracalvarial region of scid mice by a primitive stem cell population (hOMSC) derived from the lamina propria of the human oral mucosa and gingiva. MATERIAL AND METHODS: Fibrin-hOMSC constructs were cultured for 14 days at which time point they were analysed for the expression of osteoblastic/cementoblastic markers and implanted between the skin and calvaria bones into scid mice. After 8 weeks, the animals were sacrificed and the implantation sites analysed. RESULTS: Two-week-old cultures of fibrin-hOMSC constructs expressed osteogenic/cementogenic markers at the gene level. Macroscopic and radiographic examinations revealed mineralized masses at the implantation sites of fibrin-hOMSC constructs. Histology, histochemistry and immunofluorescence showed mineralized masses consisting of avascular cellular and acellular matrices that stained positively for collagen, Ca, cementum attachment protein, cementum protein 1, bone sialoprotein, alkaline phosphatase, osteocalcin, amelogenin and ameloblastin. Positive anti-human nuclear antigen indicated the human origin of the cells. Atomic force microscopy depicted long prismatic structures organized in lamellar aggregates. CONCLUSIONS: Within the limitation of this study, the results indicate for the first time that fibrin-hOMSC constructs are endowed with the constitutive capacity to develop into mineralized tissues that exhibit certain similarities to cementum and bone.


Subject(s)
Bone Regeneration , Dental Cementum/physiology , Gingiva/cytology , Mouth Mucosa/cytology , Stem Cells , Alkaline Phosphatase/biosynthesis , Amelogenin/biosynthesis , Animals , Collagen/biosynthesis , Dental Cementum/metabolism , Fibrin , Humans , Integrin-Binding Sialoprotein/biosynthesis , Mice , Mice, SCID , Osteocalcin/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Proteins/metabolism , Regeneration , Stem Cell Transplantation
5.
Bull Exp Biol Med ; 155(1): 145-50, 2013 May.
Article in English | MEDLINE | ID: mdl-23667893

ABSTRACT

We studied of osteogenic differentiation of multipotent mesenchymal stromal cells from human adipose tissue. Experiments showed that 1α,25-dihydroxycalciferol is a more effective inductor of osteogenesis than dexamethasone. Comparative analysis revealed activation of gene expression for the major osteogenic markers on day 7 of culturing in a medium containing 1α,25-dihydroxycalciferol. It was found that transcription of genes encoding type 1 collagen proteins, osteopontin, osteocalcin, and bone sialoprotein peaked on day 14 in culture, while the expression of alkaline phosphatase and bone morphogenetic protein-2 genes increased over 21 days. Intensive mineralization of the extracellular matrix was observed starting from day 14 in culture. On the basis of the analysis of these data, optimal terms for osteogenic induction (day 14) and an optimal inductor (1α,25-dihydroxycalciferol) were chosen and the protocol of effective osteogenic differentiation of multipotent mesenchymal stromal cells from human adipose tissue was developed for creation of tissue-engineered bone equivalents.


Subject(s)
Adipose Tissue/cytology , Bone Density Conservation Agents/pharmacology , Calcitriol/pharmacology , Dexamethasone/pharmacology , Mesenchymal Stem Cells/physiology , Osteogenesis , Tissue Engineering , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/genetics , Anti-Inflammatory Agents/pharmacology , Bone Morphogenetic Protein 2/biosynthesis , Bone Morphogenetic Protein 2/genetics , Cell Differentiation , Collagen Type I/biosynthesis , Collagen Type I/genetics , Extracellular Matrix/metabolism , Humans , Integrin-Binding Sialoprotein/biosynthesis , Integrin-Binding Sialoprotein/genetics , Mesenchymal Stem Cells/cytology , Osteocalcin/biosynthesis , Osteocalcin/genetics , Osteopontin/biosynthesis , Osteopontin/genetics
6.
J Biol Chem ; 286(6): 4809-18, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21135100

ABSTRACT

To avoid excess accumulation of unfolded proteins in the endoplasmic reticulum (ER), eukaryotic cells have signaling pathways from the ER to the cytosol or nucleus. These processes are collectively termed the ER stress response. Double stranded RNA activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK) is a major transducer of the ER stress response and directly phosphorylates eIF2α, resulting in translational attenuation. Phosphorylated eIF2α specifically promotes the translation of the transcription factor ATF4. ATF4 plays important roles in osteoblast differentiation and bone formation. Perk(-/-) mice are reported to exhibit severe osteopenia, and the phenotypes observed in bone tissues are very similar to those of Atf4(-/-) mice. However, the involvement of the PERK-eIF2α-ATF4 signaling pathway in osteogenesis is unclear. Phosphorylated eIF2α and ATF4 protein levels were attenuated in Perk(-/-) calvariae, and the gene expression levels of osteocalcin (Ocn) and bone sialoprotein (Bsp), which are targets for ATF4, were also down-regulated. Treatment of wild-type primary osteoblasts with BMP2, which is required for osteoblast differentiation, induced ER stress, leading to an increase in ATF4 protein expression levels. In contrast, the level of ATF4 in Perk(-/-) osteoblasts was severely diminished. The results indicate that PERK signaling is required for ATF4 activation during osteoblast differentiation. Perk(-/-) osteoblasts exhibited decreased alkaline phosphatase activities and delayed mineralized nodule formation relative to wild-type cultures. These abnormalities were almost completely restored by the introduction of ATF4 into Perk(-/-) osteoblasts. Taken together, ER stress occurs during osteoblast differentiation and activates the PERK-eIF2α-ATF4 signaling pathway followed by the promotion of gene expression essential for osteogenesis, such as Ocn and Bsp.


Subject(s)
Activating Transcription Factor 4/metabolism , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/physiology , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Osteoblasts/metabolism , Unfolded Protein Response/physiology , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Calcification, Physiologic/physiology , Endoplasmic Reticulum/genetics , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation/physiology , Integrin-Binding Sialoprotein/biosynthesis , Mice , Mice, Knockout , Osteoblasts/cytology , Osteocalcin/biosynthesis , Osteogenesis/physiology , Phosphorylation/physiology , Signal Transduction/physiology , eIF-2 Kinase/genetics
7.
J Cell Biochem ; 113(7): 2432-41, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22573557

ABSTRACT

X-linked hypophosphatemic rickets (XLH) is a dominantly inherited disease characterized by renal phosphate wasting, aberrant vitamin D metabolism, and defective bone mineralization. It is known that XLH in humans and in certain mouse models is caused by inactivating mutations in PHEX/Phex (phosphate-regulating gene with homologies to endopeptidases on the X chromosome). By a genome-wide N-ethyl-N-nitrosourea (ENU)-induced mutagenesis screen in mice, we identified a dominant mouse mutation that exhibits the classic clinical manifestations of XLH, including growth retardation, skeletal abnormalities (rickets/osteomalacia), hypophosphatemia, and increased serum alkaline phosphatase (ALP) levels. Mapping and sequencing revealed that these mice carry a point mutation in exon 14 of the Phex gene that introduces a stop codon at amino acid 496 of the coding sequence (Phex(Jrt) also published as Phex(K496X) [Ichikawa et al., 2012]). Fgf23 mRNA expression as well as that of osteocalcin, bone sialoprotein, and matrix extracellular phosphoglycoprotein was upregulated in male mutant long bone, but that of sclerostin was unaffected. Although Phex mRNA is expressed in bone from mutant hemizygous male mice (Phex(Jrt)/Y mice), no Phex protein was detected in immunoblots of femoral bone protein. Stromal cultures from mutant bone marrow were indistinguishable from those of wild-type mice with respect to differentiation and mineralization. The ability of Phex(Jrt)/Y osteoblasts to mineralize and the altered expression levels of matrix proteins compared with the well-studied Hyp mice makes it a unique model with which to further explore the clinical manifestations of XLH and its link to FGF23 as well as to evaluate potential new therapeutic strategies.


Subject(s)
Bone and Bones/pathology , Disease Models, Animal , Familial Hypophosphatemic Rickets , Genetic Diseases, X-Linked , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Point Mutation , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Bone Marrow Cells , Bone and Bones/metabolism , Calcification, Physiologic/genetics , Calcification, Physiologic/physiology , Cells, Cultured , Chromosome Mapping , Ethylnitrosourea , Extracellular Matrix Proteins/biosynthesis , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/metabolism , Familial Hypophosphatemic Rickets/pathology , Female , Fibroblast Growth Factor-23 , Glycoproteins/biosynthesis , Integrin-Binding Sialoprotein/biosynthesis , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mutagens/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/biosynthesis , Phosphoproteins/biosynthesis , RNA, Messenger/biosynthesis , Sequence Analysis, DNA , Stromal Cells
8.
Biochem Biophys Res Commun ; 424(3): 439-45, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22771798

ABSTRACT

L-type voltage-dependent Ca(2+) channels (VDCC(L)) play an important role in the maintenance of intracellular calcium homeostasis, and influence multiple cellular processes. They have been confirmed to contribute to the functional activities of osteoblasts. Recently, VDCC(L) expression was reported in mesenchymal stem cells (MSCs), but the role of VDCC(L) in MSCs is still undetermined. The aim of this study was to determine whether VDCC(L) may be regarded as a new regulator in the proliferation and osteogenic differentiation of rat MSC (rMSCs). In this study, we examined functional Ca(2+) currents (I(Ca)) and mRNA expression of VDCC(L) in rMSCs, and then suppressed VDCC(L) using nifedipine (Nif), a VDCC(L) blocker, to investigate its role in rMSCs. The proliferation and osteogenic differentiation of MSCs were analyzed by MTT, flow cytometry, alkaline phosphatase (ALP), Alizarin Red S staining, RT-PCR, and real-time PCR assays. We found that Nif exerts antiproliferative and apoptosis-inducing effects on rMSCs. ALP activity and mineralized nodules were significantly decreased after Nif treatment. Moreover, the mRNA levels of the osteogenic markers, osteocalcin (OCN), bone sialoprotein (BSP), and runt-related transcription factor 2 (Runx2), were also down-regulated. In addition, we transfected α1C-siRNA into the cells to further confirm the role of VDCC(L) in rMSCs, and a similar effect on osteogenesis was found. These results suggest that VDCC(L) plays a crucial role in the proliferation and osteogenic differentiation of rMSCs.


Subject(s)
Bone Marrow Cells/cytology , Calcium Channels, L-Type/physiology , Cell Differentiation/physiology , Cell Proliferation , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Animals , Calcium Channels, L-Type/genetics , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/biosynthesis , Integrin-Binding Sialoprotein/biosynthesis , Osteocalcin/biosynthesis , Osteogenesis/genetics , Rats
9.
Biochem Biophys Res Commun ; 425(4): 924-30, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22906741

ABSTRACT

In this study, we begin to investigate the underlying mechanism of leptin-induced vascular calcification. We found that treatment of cultured bovine aortic smooth muscle cells (BASMCs) with leptin (0.5-4 µg/ml) induced osteoblast differentiation in a dose-dependent manner. Furthermore, we found that leptin significantly increased the mRNA expression of osteopontin and bone sialoprotein, while down-regulating matrix gla protein (MGP) expression in BASMCs. Key factors implicated in osteoblast differentiation, including members of the Wnt signaling pathway, were examined. Exposure to leptin enhanced phosphorylation of GSK-3ß on serine-9 thereby inhibiting activity and promoting the nuclear accumulation of ß-catenin. Transfection of BASMCs with an adenovirus that expressed constitutively active GSK-3ß (Ad-GSK-3ß S9A) resulted in a >2-fold increase in GSK-3ß activity and a significant decrease in leptin-induced alkaline phosphatase (ALP) activity. In addition, qRT-PCR analysis showed that GSK-3ß activation resulted in a significant decrease in the expression of osteopontin and bone sialoprotein, but a marked increase in MGP mRNA expression. When taken together, our results suggest a mechanism by which leptin promotes osteoblast differentiation and vascular calcification in vivo.


Subject(s)
Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Leptin/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Osteoblasts/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Calcium-Binding Proteins/antagonists & inhibitors , Cattle , Cell Nucleus/metabolism , Cells, Cultured , Extracellular Matrix Proteins/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Integrin-Binding Sialoprotein/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Osteoblasts/cytology , Osteopontin/biosynthesis , beta Catenin/metabolism , Matrix Gla Protein
10.
Stem Cells ; 29(2): 206-16, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21732479

ABSTRACT

Reprogramming somatic cells into an ESC-like state, or induced pluripotent stem (iPS) cells, has emerged as a promising new venue for customized cell therapies. In this study, we performed directed differentiation to assess the ability of murine iPS cells to differentiate into bone, cartilage, and fat in vitro and to maintain an osteoblast phenotype on a scaffold in vitro and in vivo. Embryoid bodies derived from murine iPS cells were cultured in differentiation medium for 8­12 weeks. Differentiation was assessed by lineage-specific morphology, gene expression, histological stain, and immunostaining to detect matrix deposition. After 12 weeks of expansion, iPS-derived osteoblasts were seeded in a gelfoam matrix followed by subcutaneous implantation in syngenic imprinting control region (ICR) mice. Implants were harvested at 12 weeks, histological analyses of cell and mineral and matrix content were performed. Differentiation of iPS cells into mesenchymal lineages of bone, cartilage, and fat was confirmed by morphology and expression of lineage-specific genes. Isolated implants of iPS cell-derived osteoblasts expressed matrices characteristic of bone, including osteocalcin and bone sialoprotein. Implants were also stained with alizarin red and von Kossa, demonstrating mineralization and persistence of an osteoblast phenotype. Recruitment of vasculature and microvascularization of the implant was also detected. Taken together, these data demonstrate functional osteoblast differentiation from iPS cells both in vitro and in vivo and reveal a source of cells, which merit evaluation for their potential uses in orthopedic medicine and understanding of molecular mechanisms of orthopedic disease.


Subject(s)
Calcification, Physiologic , Induced Pluripotent Stem Cells/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Animals , Cell Differentiation , Cells, Cultured , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/cytology , Integrin-Binding Sialoprotein/biosynthesis , Mice , Mice, Inbred ICR , Mice, Nude , Osteoblasts/cytology , Osteocalcin/biosynthesis , Phenotype , Tissue Scaffolds
11.
Cells Tissues Organs ; 195(3): 244-51, 2012.
Article in English | MEDLINE | ID: mdl-21494024

ABSTRACT

One of the effects observed during several screening studies for osteocompatibility in vitro was that cells derived from the upper and lower jaw exhibited distinct differences regarding proliferation. Therefore, the aim of this study was to examine systematically whether a single osteoblast possesses abilities which are specific to the upper or lower jaw. Both human maxillary and mandibular bone samples without any clinical or radiographic evidence of pathology were obtained from 4 male donors aged between 40 and 45 years. Cells were cultured for up to 25 days to investigate in vitro development. Total and apoptotic cell numbers were estimated by image analysis. Cells were identified as bone-like cells using immunocytochemical determination of bone sialoprotein (BSP) and osteocalcin expression. The number of healthy cells was significantly higher for cells of the lower jaw compared to those of the upper jaw. The number of apoptotic cells showed an inverse pattern. The expression pattern of osseo-inductive BSP correlated with the proliferation rate of the cells. The pattern of osteocalcin expression was related to the number of apoptotic cells. Our findings are new but were anticipated regarding the well-known differences in the healing process around implants in the lower jaw versus the upper jaw. Additionally, a relationship between our results and some diseases of the lower/upper jaw seems obvious. Future work on cell responses to biomaterials should define the origin of the cells more precisely.


Subject(s)
Integrin-Binding Sialoprotein/biosynthesis , Mandible/metabolism , Osteocalcin/biosynthesis , Adult , Apoptosis/physiology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Humans , Immunohistochemistry , Integrin-Binding Sialoprotein/metabolism , Male , Mandible/cytology , Middle Aged , Osteocalcin/metabolism
12.
J Clin Periodontol ; 39(6): 556-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22512503

ABSTRACT

AIM: To investigate Glial cell line-derived neurotrophic factor (GDNF) expression in normal and wounded rat periodontal ligament (PDL) and the effects of GDNF on human PDL cells (HPDLCs) migration and extracellular matrix expression in HPDLCs. MATERIAL AND METHODS: The expression of GDNF and GDNF receptors was examined by immunocyto/histochemical analyses. Gene expression in HPDLCs treated with GDNF, interleukin-1 beta (IL-1ß), or tumour necrosis factor-alpha (TNF-α) was quantified by quantitative RT-PCR (qRT-PCR). In addition, we examined the migratory effect of GDNF on HPDLCs. RESULTS: GDNF was expressed in normal rat PDL and cultured HPDLCs. HPDLCs also expressed GDNF receptors. In wounded rat PDL, GDNF expression was up-regulated. QRT-PCR analysis revealed that IL-1ß and TNF-α significantly increased the expression of GDNF in HPDLCs. Furthermore, GDNF induced migration of HPDLCs, which was blocked by pre-treatment with the peptide including Arg-Gly-Asp (RGD) sequence, or neutralizing antibodies against integrin αVß3 or GDNF. Also, GDNF up-regulated expression of bone sialoprotein (BSP) and fibronectin in HPDLCs. CONCLUSIONS: GDNF expression is increased in rat wounded PDL tissue and HPDLCs treated with pro-inflammatory cytokines. GDNF enhances the expression of BSP and fibronectin, and migration in an RGD-dependent manner via the integrin αVß3. These findings suggest that GDNF may contribute to wound healing in PDL tissue.


Subject(s)
Alveolar Bone Loss/metabolism , Extracellular Matrix Proteins/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Adult , Animals , Cell Adhesion , Cell Movement/drug effects , Cells, Cultured , Extracellular Matrix Proteins/genetics , Female , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Integrin-Binding Sialoprotein/biosynthesis , Integrin-Binding Sialoprotein/genetics , Interleukin-1beta/pharmacology , Male , Oligopeptides/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/pathology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Wound Healing/physiology , Young Adult
13.
Int Endod J ; 45(12): 1080-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22747502

ABSTRACT

AIM: To compare cells from normal and inflamed human dental pulps regarding the presence of stem cells, their proliferation and differentiation potential. METHODOLOGY: Human dental pulp stem cells (hDPSCs) were isolated from normal (DPSC-N) and inflamed dental pulps (DPSC-I). They were compared in respect to proliferation (MTT assay), morphology and STRO-1 expression. STRO-1-positive cells were subject to proliferation (MTT and CFU counting) and morphological analyses and then submitted to odonto-osteogenic, adipogenic and condrogenic differentiation. Differentiated cells were evaluated concerning morphology and the expression, by qRT-PCR, of BSP, LPL and SOX-9 genes. The amount of mineralized matrix produced after odonto-osteogenic differentiation was compared with quantitative Alizarin Red staining. RESULTS: No difference was observed in the morphology and in the proliferation rate of DPSC-N and DPSC-I either before or after separation of STRO-1-positive cells. These cells represented 0.46% (±0.14) and 0.43% (±0.19) of the cell population from normal and inflamed dental pulps, respectively. Both DPSC-N and DPSC-I were capable of differentiating under the three assayed conditions and presented similar patterns for BSP, LPL and SOX-9 expression. Mineralized matrix production was also compatible. In all the quantitative experiments, differences were found between cells from each patient, either from normal or from inflamed pulps. Nonetheless, there was no statistical difference between these two groups. CONCLUSION: The morphology, proliferation rate and differentiation potential of DPSC-I were similar to the observed in DPSC-N, thus demonstrating that the inflammatory process did not affect the stem cell properties that were assessed.


Subject(s)
Dental Pulp/cytology , Mesenchymal Stem Cells , Pulpitis/pathology , Adipogenesis , Adolescent , Adult , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrogenesis , Flow Cytometry , Humans , Immunophenotyping , Integrin-Binding Sialoprotein/biosynthesis , Lipoprotein Lipase/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Osteogenesis , Regeneration , SOX9 Transcription Factor/biosynthesis , Young Adult
14.
Biochem Biophys Res Commun ; 412(2): 273-8, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21820411

ABSTRACT

Zinc is one of the trace elements which induce the proliferation and the differentiation of the osteoblast. In the previous study, we found that zinc ions (Zn(2+) ion)-releasing titanium implants had excellent bone fixation using a rabbit femurs model. In this study, we isolated the Zn(2+) ions (eluted Zn(2+) ion; EZ) released from the implant surface, and evaluated the effect of EZ on the osteogenesis of human bone marrow-derived mesenchymal cells (hBMCs). In the result, it was found that the EZ stimulated cell viability, osteoblast marker gene (type I collagen, osteocalcin (OC), alkaline phosphatase (ALP) and bone sialoprotein (BSP)) expressions and calcium deposition in hBMCs.


Subject(s)
Bone Regeneration/drug effects , Osteogenesis/drug effects , Zinc/metabolism , Alkaline Phosphatase/biosynthesis , Animals , Bone Marrow Cells/drug effects , Cations, Divalent/metabolism , Cations, Divalent/pharmacology , Cells, Cultured , Collagen Type I/biosynthesis , Drug Implants/metabolism , Drug Implants/pharmacology , Femur , Integrin-Binding Sialoprotein/biosynthesis , Mesoderm/drug effects , Osteocalcin/biosynthesis , RNA, Messenger/biosynthesis , Rabbits , Surface Properties , Titanium , Zinc/pharmacology
15.
Biotechnol Bioeng ; 108(11): 2727-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21656707

ABSTRACT

Osteoconductive materials play a critical role in promoting integration with surrounding bone tissue and resultant bone repair in vivo. However, the impact of 3D osteoconductive substrates coupled with soluble signals on progenitor cell differentiation is not clear. In this study, we investigated the influence of bone morphogenetic protein-2 (BMP-2) concentration on the osteogenic differentiation of human mesenchymal stem cells (hMSCs) when seeded in carbonated apatite-coated polymer scaffolds. Mineralized scaffolds were more hydrophilic and adsorbed more BMP-2 compared to non-mineralized scaffolds. Changes in alkaline phosphatase (ALP) activity within stimulated hMSCs were dependent on the dose of BMP-2 and the scaffold composition. We detected more cell-secreted calcium on mineralized scaffolds at all time points, and higher BMP-2 concentrations resulted in increased ALP and calcium levels. RUNX2 and IBSP gene expression within hMSCs was affected by both substrate and soluble signals, SP7 by soluble factors, and SPARC by substrate-mediated cues. The present data indicate that a combination of apatite and BMP-2 do not simply enhance the osteogenic response of hMSCs, but act through multiple pathways that may be both substrate- and growth factor-mediated. Thus, multiple signaling strategies will likely be necessary to achieve optimal bone regeneration.


Subject(s)
Apatites/metabolism , Bone Morphogenetic Protein 2/metabolism , Mesenchymal Stem Cells/physiology , Osteogenesis , Alkaline Phosphatase/metabolism , Calcium/metabolism , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/biosynthesis , Gene Expression Profiling , Humans , Integrin-Binding Sialoprotein/biosynthesis , Mesenchymal Stem Cells/drug effects , Osteonectin/biosynthesis , Sp7 Transcription Factor , Transcription Factors/biosynthesis
16.
Connect Tissue Res ; 52(5): 393-400, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21405979

ABSTRACT

In this study, we try to compare the histological characteristics and the odontogenic capability of apical tissues (AT) at different root development stages of rat molar teeth. AT of mandibular first molars from 8-day-old, 21-day-old, and 35-day-old Sprague-Dawley rats were selected as being representative of root-initiating, root-forming, and root-completing stages, respectively. Cell counting, flow cytometry assays, alkaline phosphatase activity, alizarin red staining, and reverse transcription polymerase chain reaction were performed to assess the proliferation and mineralization potential of apical tissue cells at different stages of root development in vitro. In vivo transplantation of apical tissue cells combined with ceramic bovine bone was used to characterize the differentiation capacity. It was shown that there was a structurally and functionally dynamic change in the apical tissue of developing tooth root of rats, of which the unique developmental potential will reduce gradually with the ending up of root development. The AT of root-initiating and root-forming stage exhibited much higher proliferation and tissue-regenerative capacity than those of root-completing stage. Our present results indicate that the apical tissue, with the sustainable developmental ability throughout almost the whole process of tooth development, can yet be regarded as a competent candidate source for root/periodontal tissues regeneration.


Subject(s)
Molar/growth & development , Odontogenesis/physiology , Tooth Apex/growth & development , Tooth Root/growth & development , Alkaline Phosphatase/metabolism , Animals , Cattle , Cell Proliferation , Cells, Cultured , Dental Enamel Proteins/biosynthesis , Gene Expression , Integrin-Binding Sialoprotein/biosynthesis , Molar/cytology , Osteocalcin/biosynthesis , Osteopontin/biosynthesis , Periodontium/growth & development , Rats , Rats, Sprague-Dawley , Regeneration , Tooth Apex/transplantation , Tooth Calcification , Tooth Root/cytology
17.
Am J Orthod Dentofacial Orthop ; 139(4 Suppl): S83-101, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435543

ABSTRACT

BACKGROUND AND PURPOSE: The aim of this study was to test if corticotomy-induced osteoclastogenesis and bone remodeling underlie orthodontic tooth movement and how selective alveolar decortication enhances the rate of tooth movement. MATERIALS AND METHODS: A total of 114 Sprague-Dawley rats were included in 3 treatment groups: selective alveolar decortication alone (SADc); tooth movement alone (TM); and "combined" therapy (SADc + TM). Surgery was performed around the buccal and palatal aspects of the left maxillary first molar tooth and included 5 decortication dots on each side. Tooth movement was performed on the first molar using a 25-g Sentalloy spring. Measurements were done at baseline (day 0: no treatment rendered) and on days 3, 7, 14, 21, 28 and 42. Microcomputed tomography, Faxitron analyses, and quantitative real-time polymerase chain reaction (q-PCR) of expressed mRNAs were used to assess changes. RESULTS: The combined group showed increased tooth movement (P = 0.04) at 7 days compared with the tooth movement group with significantly decreased bone volume (62%; P = 0.016) and bone mineral content (63%; P = 0.015). RNA markers of osteoclastic cells and key osteoclastic regulators (M-CSF [macrophage colony-stimulating factor], RANKL [receptor activator of nuclear factor kappa-B ligand], OPG [osteoprotegerin], calcitonin receptor [CTR], TRACP-5b [tartrate-resistant acid phosphatase 5b], cathepsin K [Ctsk]) all showed expression indicating increased osteoclastogenesis in the combined group. RNA markers of osteoblastic cells (OPN [osteopontin], BSP [bone sialoprotein], OCN [osteocalcin]) also showed increased anabolic activity in response to the combination of alveolar decortication and tooth movement. CONCLUSIONS: The data suggest that the alveolar decortication enhances the rate of tooth movement during the initial tooth displacement phase; this results in a coupled mechanism of bone resorption and bone formation during the earlier stages of treatment, and this mechanism underlies the rapid orthodontic tooth movement.


Subject(s)
Alveolar Process/surgery , Bone Remodeling , Dental Stress Analysis/methods , Tooth Movement Techniques/methods , Alveolar Process/diagnostic imaging , Alveolar Process/pathology , Animals , Bone Density , Bone Remodeling/genetics , Cathepsin K/biosynthesis , Integrin-Binding Sialoprotein/biosynthesis , Macrophage Colony-Stimulating Factor/biosynthesis , Maxilla , Osteoblasts/metabolism , Osteocalcin/biosynthesis , Osteoclasts/metabolism , Osteopontin/biosynthesis , Osteoprotegerin/biosynthesis , Periodontal Ligament/physiology , Polymerase Chain Reaction , RANK Ligand/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Calcitonin/biosynthesis , X-Ray Microtomography
18.
Cells ; 10(6)2021 05 24.
Article in English | MEDLINE | ID: mdl-34073955

ABSTRACT

Bone sialoprotein (BSP) has become a target in breast cancer research as it is associated with tumor progression and metastasis. The mechanisms underlying the regulation of BSP expression have been largely elusive. Given that BSP is involved in the homing of cancer cells in bone metastatic niches, we addressed regulatory effects of proteolytic cleavage and extracellular matrix components on BSP expression and distribution in cell culture models. Therefore, MDA-MB-231 human breast cancer cells were kept in 2D and 3D spheroid cultures and exposed to basement membrane extract in the presence or absence of matrix metalloproteinase 9 or the non-polar protease, dispase. Confocal imaging of immunofluorescence samples stained with different antibodies against human BSP demonstrated a strong inducing effect of basement membrane extract on anti-BSP immunofluorescence. Similarly, protease incubation led to acute upregulation of anti-BSP immunofluorescence signals, which was blocked by cycloheximide, suggesting de novo formation of BSP. In summary, our data show that extracellular matrix components play an important function in regulating BSP expression and hint at mechanisms for the formation of bone-associated metastasis in breast cancer that might involve local control of BSP levels by extracellular matrix degradation and release of growth factors.


Subject(s)
Breast Neoplasms/metabolism , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Integrin-Binding Sialoprotein/biosynthesis , Neoplasm Proteins/biosynthesis , Breast Neoplasms/pathology , Cell Line, Tumor , Extracellular Matrix/pathology , Female , Humans
19.
J Periodontal Res ; 45(5): 643-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20572923

ABSTRACT

BACKGROUND AND OBJECTIVE: Enamel sheath protein (ESP) is involved in the construction of the enamel sheath during tooth development. The 17 kDa ESP is a one-step cleavage product processed by proteolysis from the N-terminal side of sheathlin (ameloblastin/amelin), one of the porcine enamel matrix proteins. Enamel sheath protein exhibits periodontal ligament and cementum regeneration activity in a buccal dehiscence model in dogs, and promotes the cytodifferentiation of cultured human periodontal ligament (HPDL) cells. The aim of this study was to determine the peptide segment on the C-terminal side sequence of the human ESP that possesses a cytodifferentiation activity on cultured HPDL cells. MATERIAL AND METHODS: The peptides synthesized on the basis of human ESP C-terminal side sequence were tested for their ability to increase the alkaline phosphatase (ALP) and mineralization activity of cultured HPDL cells. The expressions of osteocalcin, osteopontin and bone sialoprotein were measured by semi-quantitative PCR and therefore were determined to be specific indicators of mineralized tissue differentiation. RESULTS: Multiple synthetic peptides from the human ESP increased the ALP activity and stimulated matrix mineralization in long-term cultures of HPDL cells. Semi-quantitative PCR demonstrated the osteocalcin, osteopontin and bone sialoprotein expressions to increase relative to the control values. The peptide SDKPPKPELPGVDF had the strongest cytodifferentiation activity among all the synthetic peptides tested. CONCLUSION: A specific peptide sequence derived from the C-terminal side of the human ESP promotes the cytodifferentiation and mineralization activity of HPDL cells in a cell culture system.


Subject(s)
Dental Enamel Proteins/chemical synthesis , Dental Enamel Proteins/physiology , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Alkaline Phosphatase/biosynthesis , Amino Acid Sequence , Animals , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Cementogenesis/drug effects , Cementogenesis/physiology , Dental Enamel Proteins/chemistry , Dental Enamel Proteins/pharmacology , Humans , Integrin-Binding Sialoprotein/biosynthesis , Mice , Molecular Sequence Data , Osteocalcin/biosynthesis , Osteopontin/biosynthesis , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Periodontal Ligament/metabolism , Regeneration/drug effects , Regeneration/physiology
20.
J Periodontal Res ; 45(5): 602-11, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20546115

ABSTRACT

BACKGROUND AND OBJECTIVE: Amelogenins are hydrophobic proteins that are the major component of developing enamel. Enamel matrix derivative has been used for periodontal regeneration. Bone sialoprotein is an early phenotypic marker of osteoblast differentiation. In this study, we examined the ability of porcine amelogenins to regulate bone sialoprotein transcription. MATERIAL AND METHODS: To determine the molecular basis of the transcriptional regulation of the bone sialoprotein gene by amelogenins, we conducted northern hybridization, transient transfection analyses and gel mobility shift assays using the osteoblast-like ROS 17/2.8 cells. RESULTS: Amelogenins (100 ng/mL) up-regulated bone sialoprotein mRNA at 3 h, with maximal mRNA expression occurring at 12 h (25 and 20 kDa) and 6 h (13 and 6 kDa). Amelogenins (100 ng/mL, 12 h) increased luciferase activities in pLUC3 (nucleotides -116 to +60), and 6 kDa amelogenin up-regulated pLUC4 (nucleotides -425 to +60) activity. The tyrosine kinase inhibitor inhibited amelogenin-induced luciferase activities, whereas the protein kinase A inhibitor abolished 25 kDa amelogenin-induced bone sialoprotein transcription. The effects of amelogenins were abrogated by 2-bp mutations in the fibroblast growth factor 2 response element (FRE). Gel-shift assays with radiolabeled FRE, homeodomain-protein binding site (HOX) and transforming growth factor-beta1 activation element (TAE) double-strand oligonucleotides revealed increased binding of nuclear proteins from amelogenin-stimulated ROS 17/2.8 cells at 3 h (25 and 13 kDa) and 6 h (20 and 6 kDa). CONCLUSION: These results demonstrate that porcine 25 kDa amelogenin and its proteolytic derivatives stimulate bone sialoprotein transcription by targeting FRE, HOX and TAE in the bone sialoprotein gene promoter, and that full-length amelogenin and amelogenin cleavage products are able to regulate bone sialoprotein transcription via different signaling pathways.


Subject(s)
Amelogenin/pharmacology , Integrin-Binding Sialoprotein/biosynthesis , Promoter Regions, Genetic/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Amelogenin/metabolism , Animals , Binding Sites , Blotting, Northern , Cell Line , DNA Probes , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic/drug effects , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Integrin-Binding Sialoprotein/genetics , Luciferases/metabolism , Mutagenesis, Site-Directed , Osteoblasts/drug effects , Osteoblasts/metabolism , Rats , Response Elements/drug effects , Swine , Transfection , Transforming Growth Factor beta1/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL