Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 824
Filter
1.
J Biomed Mater Res A ; 109(5): 682-694, 2021 05.
Article in English | MEDLINE | ID: mdl-32608088

ABSTRACT

This study evaluated the response of a nano-hydroxyapatite coating implant through gene expression analysis (runt-related transcription factor 2 (Runx2), alkaline phosphatase (Alp), osteopontin (Opn), osteocalcin (Oc), receptor activator of nuclear factor-kappa B (Rank), receptor activator of nuclear factor-kappa B ligand (Rank-L), and osteoprotegerin (Opg)). Three-dimensional evaluation (percent bone volume (BV/TV); percent intersection surface (BIC); bone surface/volume ratio (BS/BV); and total porosity (To.Po)) were also analyzed. Mini implants were surgically placed in tibias of both healthy and diabetic rats. The animals were euthanized at 7 and 30 days. Evaluating all factors the relative expression of Rank showed that NANO surface presented the best results at 7 days (diabetic rats). Furthermore the levels of Runx2, Alp, Oc, and Opn suggest an increase in osteoblasts proliferation, especially in early stages of osseointegration. %BIC in healthy and diabetic (7 days) depicted statistically significant differences for NANO group. BV/TV, BS/BV and To.Po demonstrated higher values for NANO group in all evaluated time point and irrespective of systemic condition, but BS/BV 30 days (healthy rat) and 7 and 30 days (diabetic rat). Microtomographic and gene expression analyses have shown the benefits of nano-hydroxyapatite coated implants in promoting new bone formation in diabetic rats.


Subject(s)
Bone Regeneration/drug effects , Coated Materials, Biocompatible/pharmacology , Durapatite/pharmacology , Gene Expression Regulation/drug effects , Implants, Experimental , Nanoparticles , Osteogenesis/drug effects , X-Ray Microtomography , Animals , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Diabetes Mellitus, Experimental , Durapatite/therapeutic use , Male , Microscopy, Electron, Scanning , Nanoparticles/therapeutic use , Osseointegration , Osteocalcin/biosynthesis , Osteocalcin/genetics , Osteogenesis/genetics , Osteopontin/biosynthesis , Osteopontin/genetics , Osteoprotegerin/biosynthesis , Osteoprotegerin/genetics , RANK Ligand/biosynthesis , RANK Ligand/genetics , Rats , Rats, Wistar , Tibia/surgery
2.
Mediators Inflamm ; 2020: 2793804, 2020.
Article in English | MEDLINE | ID: mdl-33204216

ABSTRACT

Although postmenopausal osteoporosis often occurs concurrently with diabetes, little is known about interactions between estrogen deficiency and hyperglycemia in the skeletal system. In the present study, the effects of estrogen deficiency on the development of biochemical, microstructural, and mechanical changes induced by streptozotocin-induced diabetes mellitus (DM) in the rat skeletal system were investigated. The experiments were carried out on nonovariectomized (NOVX) and ovariectomized (OVX) control and diabetic mature female Wistar rats. Serum levels of bone turnover markers (CTX-I and osteocalcin) and 23 cytokines, bone mass and mineralization, histomorphometric parameters, and mechanical properties of cancellous and compact bone were determined. The results were subjected to two-way ANOVA and principal component analysis (PCA). Estrogen deficiency induced osteoporotic changes, with increased bone resorption and formation, and worsening of microstructure (femoral metaphyseal BV/TV decreased by 13.0%) and mechanical properties of cancellous bone (the maximum load in the proximal tibial metaphysis decreased by 34.2%). DM in both the NOVX and OVX rats decreased bone mass, increased bone resorption and decreased bone formation, and worsened cancellous bone microarchitecture (for example, the femoral metaphyseal BV/TV decreased by 17.3% and 18.1%, respectively, in relation to the NOVX controls) and strength (the maximum load in the proximal tibial metaphysis decreased by 35.4% and 48.1%, respectively, in relation to the NOVX controls). Only in the diabetic rats, profound increases in some cytokine levels were noted. In conclusion, the changes induced by DM in female rats were only slightly intensified by estrogen deficiency. Despite similar effects on bone microstructure and strength, the influence of DM on the skeletal system was based on more profound systemic homeostasis changes than those induced by estrogen deficiency.


Subject(s)
Bone and Bones/pathology , Diabetes Mellitus, Type 1/physiopathology , Estrogens/deficiency , Animals , Bone and Bones/metabolism , Collagen Type I , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Estrogens/metabolism , Female , Femur/physiopathology , Homeostasis , Osteocalcin/biosynthesis , Ovary/surgery , Peptide Fragments , Principal Component Analysis , Rats , Rats, Wistar , Stress, Mechanical
3.
J Cell Mol Med ; 24(9): 4944-4955, 2020 05.
Article in English | MEDLINE | ID: mdl-32198976

ABSTRACT

Bone defects resulting from non-union fractures or tumour resections are common clinical problems. Long non-coding RNAs (lncRNAs) are reported to play vital roles in stem cell differentiation. The aim of this study was to elucidate the role of lncRNA-H19 in osteogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs). Following the establishment of an osteogenic differentiation model in rats, the expression of H19, microRNA-149 (miR-149) and stromal cell-derived factor-1 (SDF-1) was measured by RT-qPCR. Thereafter, BMMSCs were isolated from rats and treated with a series of mimic, inhibitor or siRNA. SDF-1 expression, alkaline phosphatase (ALP) activity and osteocalcin (OCN) content were detected. The mineralized and calcified nodules were assessed by alizarin red S and Von Kossa staining. BMMSC surface markers were detected by flow cytometry. Western blot analysis was used to measure the expression of ALP, OCN, runt-related transcription factor 2 (RUNX2) and osterix (OSX) proteins. Lastly, dual-luciferase reporter gene assay and RNA immunoprecipitation were applied to verify the relationship of H19, miR-149 and SDF-1. Overexpressed H19 and SDF-1 and poorly expressed miR-149 were found in rats with osteogenic differentiation. H19 increased SDF-1 expression by binding to miR-149. H19 enhanced ALP activity, OCN content, calcium deposit and ALP, OCN, RUNX2 and OSX protein expression of BMMSCS by up-regulating SDF-1 via binding to miR-149. Taken together, up-regulated H19 could promote the osteogenic differentiation of BMMSCs by increasing SDF-1 via miR-149.


Subject(s)
Bone Marrow Cells/cytology , Chemokine CXCL12/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Alkaline Phosphatase/metabolism , Animals , Bone Regeneration , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Genes, Reporter , Male , Osteocalcin/biosynthesis , Osteogenesis , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism , Transfection , Up-Regulation
4.
Biol Trace Elem Res ; 193(1): 195-203, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30887282

ABSTRACT

The gap junction protein plays an important role in the bone formation and alteration of these proteins leading to cause bone development. Aim to determine the effects of different concentration of fluoride on gap-junctional intercellular communication (GJIC) related genes and proteins in the rats' osteoblast cells. We treated the osteoblast cells with various concentrations (0, 0.01, 0.1, 0.5, and 1.0 mM) NaF for 24 and 72 h. We used the scrape loading and dye transfer technique to research the intracellular connectivity. Moreover, the mRNA expression levels of connexin 43 (Cx43), connexin45 (Cx45), collagen I, and osteocalcin (OCN) were analyzed by qRT-PCR, the protein expression levels of connexin43 (Cx43) were analyzed by western blotting and immunofluorescence. Our results suggested that the osteoblast proliferations were decreased in the 0.5 and 1 mM NaF groups, after 24 and 72 treatments. The scrape loading and dye transfer experiment showed that the GJIC were increased in the 0.01 mM NaF group and decreased in the 0.5 and 1 mM NaF groups. In addition, the mRNA expressions of Cx43, Cx45, and OCN, and the protein expressions of Cx43 were increased in the 0.01 mM NaF group and decreased in the 0.5 and 1 mM NaF groups. In summary, these results suggest that the low concentration NaF is good for the GJIC, but the high concentration NaF damages the GJIC.


Subject(s)
Cell Communication/drug effects , Fluorides/pharmacology , Gap Junctions/metabolism , Osteoblasts/metabolism , Animals , Cells, Cultured , Connexin 43/biosynthesis , Connexins/biosynthesis , Gene Expression Regulation/drug effects , Osteocalcin/biosynthesis , Rats
5.
J Tissue Eng Regen Med ; 13(12): 2256-2265, 2019 12.
Article in English | MEDLINE | ID: mdl-31677246

ABSTRACT

BACKGROUND: Calcium ions (Ca2+ ) influence natural bone healing, and calcium is frequently used in bone tissue engineering scaffolds and cements. Scaffolds can also incorporate gene delivery systems to further promote osteoblast differentiation. Thus, our goal was to identify if Ca2+ concentration affects the transfection of bone marrow stromal cells because these cells play a major role in bone healing and can infiltrate gene-activated scaffolds designed to promote bone growth. METHODS: Bone marrow-derived mesenchymal stem cells (BMSCs) were cultured in media with Ca2+ concentrations ranging from 0 to 20 mM and transfected with polyethyleneimine-plasmid DNA (PEI-pDNA) complexes. Cell viability and transfection efficiency were determined using MTS assays and flow cytometry, respectively. PEI-pDNA complex localization in BMSCs was assessed using fluorescence microscopy. To determine BMSC differentiation, messenger RNA (mRNA) for osteocalcin and CBFA1 was quantified using real time-polymerase chain reaction (PCR). Calcium deposition was qualitatively assessed after three and 14 days using Alizarin Red staining. RESULT: Our results indicate that Ca2+ levels between 8 and 12 mM positively impacted transfection of BMSCs with PEI-pDNA complexes in terms of cell viability and transfection efficiency. A Ca2+ concentration of 10 mM also increased the expression of an osteogenic gene, osteocalcin, when the cells were transfected with plasmid DNA encoding bone morphogenetic protein 2 (BMP-2). CONCLUSION: Ca2+ at a 10 mM concentration can significantly reduce toxicity and enhance transfection efficiency when combined with PEI-pDNA complexes, and this combination can be specifically applied to further enhance the differentiation of BMSCs by using the combination of polyethyleneimine-plasmid bone morphogenetic protein 2 (PEI-pBMP-2) and 10 mM Ca2+ as compared with PEI-pBMP-2 alone.


Subject(s)
Bone Marrow Cells/metabolism , Calcium/pharmacology , Osteoblasts/metabolism , Stem Cells/metabolism , Transfection , Bone Marrow Cells/cytology , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , HEK293 Cells , Humans , Osteoblasts/cytology , Osteocalcin/biosynthesis , Osteocalcin/genetics , Stem Cells/cytology , Stromal Cells/cytology , Stromal Cells/metabolism
6.
Exp Hematol ; 80: 1-10, 2019 12.
Article in English | MEDLINE | ID: mdl-31765673

ABSTRACT

The processes generating cells of adaptive immunity render them less amenable to the single cytokine signals used so effectively to regenerate myeloid cells. T-cell neogenesis begins in the bone marrow, where specific sets of late osteolineage cells govern the specification of hematopoietic cells capable of migrating to the thymus where differentiation is completed. Osteocalcin-expressing bone marrow stromal cells producing Dll4 serve as a progenitor niche enabling this T-competent cell production. Biocompatible alginate-based cryogels containing bone morphogenetic proteins (BMP-2) and the Notch ligand Dll4 were engineered to recapitulate the endogenous niche. These cryogels are highly pliable and can be injected under the skin of animals undergoing bone marrow transplantation. The result in mice is an ectopic niche fostering T-competent progenitor generation that results in improved T-cell numbers and receptor diversity. The recipients can generate neoantigen vaccine responses while having improved tolerance manifest by reduced graft-versus-host disease upon allogeneic transplant. Through emerging details of niches in the bone marrow, therapeutics more complex than those necessary for myeloid reconstitution are possible. Niche biology-guided bioengineered design offers the possibility of regenerative therapies for T lymphoid cells.


Subject(s)
Cell Engineering/methods , Lymphopoiesis , Regenerative Medicine/methods , Stem Cell Niche/physiology , T-Lymphocyte Subsets/cytology , Adaptor Proteins, Signal Transducing/pharmacology , Adult , Age Factors , Alginates , Animals , Atrophy , Australia , Awards and Prizes , Bone Marrow/physiology , Bone Morphogenetic Protein 2/pharmacology , Calcium-Binding Proteins/pharmacology , Cell Lineage , Cryogels/pharmacology , Heterografts , Humans , Mice , Osteocalcin/biosynthesis , Societies, Scientific , Stem Cell Research , T-Lymphocyte Subsets/transplantation , Thymus Gland/pathology
7.
Am J Physiol Cell Physiol ; 317(5): C932-C941, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31411920

ABSTRACT

Exosome secretion is an important paracrine way of endothelial progenitor cells (EPCs) to modulate resident endothelial cells. The osteocalcin (OCN)-expressing EPCs have been found to be increased in cardiovascular disease patients and are considered to be involved in the process of coronary atherosclerosis. Since OCN has been proven to prevent endothelial dysfunction, this study aimed to evaluate the effect of exosomes derived from OCN-overexpressed EPCs on endothelial cells. Exosomes derived from EPCs (Exos) and OCN-overexpressed EPCs (OCN-Exos) were isolated and incubated with rat aorta endothelial cells (RAOECs) with or without the inhibition of OCN receptor G protein-coupled receptor family C group 6 member A (GPRC6A). The effects of exosomes on the proliferation activity of endothelial cells were evaluated by CCK-8 assay, and the migration of endothelial cells was detected by wound healing assay. A tube formation assay was used to test the influence of exosomes on the angiogenesis performance of endothelial cells. Here, we presented that OCN was packed into Exos and was able to be transferred to the RAOECs via exosome incorporation, which was increased in OCN-Exos groups. Compared with Exos, OCN-Exos had better efficiency in promoting RAOEC proliferation and migration and tube formation. The promoting effects were impeded after the inhibition of GPRC6A expression in RAOECs. These data suggest that exosomes from OCN-overexpressed EPCs have a beneficial regulating effect on endothelial cells, which involved enhanced OCN-GPRC6A signaling.


Subject(s)
Cell Proliferation/physiology , Endothelial Progenitor Cells/metabolism , Exosomes/metabolism , Neovascularization, Physiologic/physiology , Osteocalcin/biosynthesis , Animals , Cell Movement/physiology , Gene Expression , Osteocalcin/genetics , Rats
8.
Actual. osteol ; 15(2): 78-93, mayo - ago. 2019. ilus.
Article in Spanish | LILACS | ID: biblio-1048450

ABSTRACT

Los hallazgos osteológicos se intensi!caron en los últimos años. Se demostró que el esqueleto se comporta, además de sus funciones clásicas, como un órgano de secreción endocrina que sintetiza al menos dos hormonas: el factor de crecimiento de !broblastos 23 (FGF-23) y la osteocalcina (Ocn). La Ocn es un péptido pequeño que contiene 3 residuos de ácido glutámico. Estos residuos se carboxilan postraduccionalmente, quedando retenida en la matriz ósea. La forma decarboxilada en el primer residuo de ácido glutámico (GluOcn) fue reportada por poseer efectos biológicos; la resorción ósea es el mecanismo clave para su bioactivación. La presente revisión se centra en los conocimientos actuales sobre la función hormonal de la Ocn. A la fecha se reporta que la Ocn regularía el metabolismo energético aumentando la proliferación de células ` pancreáticas, y la secreción de insulina y de adiponectina. Sobre el músculo esquelético actuaría favoreciendo la absorción y el catabolismo de nutrientes. La función reproductiva masculina estaría regulada mediante el estímulo a las células de Leydig para sintetizar testosterona; en el desarrollo cerebral y la cognición, la Ocn aumentaría la síntesis de neurotransmisores monoaminados y disminuiría el neurotransmisor inhibidor GABA. Si bien son indispensables mayores evidencias para dilucidar los mecanismos reguladores por medio de los cuales actuaría la Ocn, los resultados enumerados en los distintos estudios experimentales establecen la importancia de este novedoso integrante molecular. Dilucidar su rol dentro de estos procesos interrelacionados en seres humanos abriría la posibilidad de utilizar a la Ocn en el tratamiento de enfermedades endocrino-metabólicas. (AU)


Osteological !ndings have intensi!ed in recent years. The skeleton behaves as an endocrine secretion organ that synthesizes at least two hormones: osteocalcin (Ocn) and !broblast growth factor 23 (FGF-23). Ocn is a small peptide that contains 3 glutamic acid residues. After translation, these residues are carboxylated to make possible its retention into the bone matrix. Decarboxylation on the !rst glutamic acid residue (GluOcn) has been reported to have biological effects. Bone resorption is the key mechanism for its bioactivation. This review focuses on current knowledge on Ocn hormonal function. It has been reported that Ocn regulates energy metabolism by increasing the proliferation of pancreatic ` cells, and the secretion of insulin and adiponectin. On the skeletal muscle, it may act by favoring the absorption and catabolism of nutrients. Male reproductive function might be regulated by stimulating Leydig cells to synthesize testosterone. Regarding brain development and cognition, Ocn would increase monoamine neurotransmitters synthesis and decrease inhibitory neurotransmitter GABA. Although more evidence is needed to elucidate the regulatory mechanisms of Ocn, different experimental studies establish the importance of this novel molecular mediator. Clarifying its role within interrelated processes in humans, might open the possibility of using Ocn in different treatments of endocrine-metabolic diseases. (AU)


Subject(s)
Animals , Osteocalcin/metabolism , Osteocalcin/therapeutic use , Skeleton/physiology , Skeleton/metabolism , Skeleton/pathology , Warfarin/therapeutic use , Cardiovascular Diseases/prevention & control , Osteocalcin/biosynthesis , Osteocalcin/chemistry , Diabetes Mellitus, Type 2/prevention & control , Endocrine System Diseases/therapy , Energy Metabolism/physiology , Insulin-Secreting Cells/physiology , Fertility , Fibroblast Growth Factors/metabolism , Genitalia, Male/metabolism , Infertility/prevention & control , Metabolic Diseases/therapy , Neoplasms/prevention & control
9.
FASEB J ; 33(6): 7331-7347, 2019 06.
Article in English | MEDLINE | ID: mdl-30884976

ABSTRACT

Osteoblasts are versatile cells involved in multiple whole-body processes, including bone formation and immune response. Secretory amounts and patterns of osteoblast-derived proteins such as osteopontin (OPN) and osteocalcin (OCN) modulate osteoblast function. However, the regulatory mechanism of OPN and OCN expression remains unknown. Here, we demonstrate that p54/p46 c-jun N-terminal kinase (JNK) inhibition suppresses matrix mineralization and OCN expression but increases OPN expression in MC3T3-E1 cells and primary osteoblasts treated with differentiation inducers, including ascorbic acid, bone morphogenic protein-2, or fibroblast growth factor 2. Preinhibition of JNK before the onset of differentiation increased the number of osteoblasts that highly express OPN but not OCN (OPN-OBs), indicating that JNK affects OPN secretory phenotype at the early stage of osteogenic differentiation. Additionally, we identified JNK2 isoform as being critically involved in OPN-OB differentiation. Microarray analysis revealed that OPN-OBs express characteristic transcription factors, cell surface markers, and cytokines, including glycoprotein hormone α2 and endothelial cell-specific molecule 1. Moreover, we found that inhibitor of DNA binding 4 is an important regulator of OPN-OB differentiation and that dual-specificity phosphatase 16, a JNK-specific phosphatase, functions as an endogenous regulator of OPN-OB induction. OPN-OB phenotype was also observed following LPS from Porphyromonas gingivalis stimulation during osteogenic differentiation. Collectively, these results suggest that the JNK-Id4 signaling axis is crucial in the control of OPN and OCN expression during osteoblastic differentiation.-Kusuyama, J., Amir, M. S., Albertson, B. G., Bandow, K., Ohnishi, T., Nakamura, T., Noguchi, K., Shima, K., Semba, I., Matsuguchi, T. JNK inactivation suppresses osteogenic differentiation, but robustly induces osteopontin expression in osteoblasts through the induction of inhibitor of DNA binding 4 (Id4).


Subject(s)
Inhibitor of Differentiation Proteins/physiology , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/physiology , Osteoblasts/metabolism , Osteogenesis/physiology , Osteopontin/biosynthesis , Animals , Cells, Cultured , Dual-Specificity Phosphatases/deficiency , Dual-Specificity Phosphatases/physiology , Gene Expression Regulation, Developmental/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/physiology , Mitogen-Activated Protein Kinase Phosphatases/deficiency , Mitogen-Activated Protein Kinase Phosphatases/physiology , Osteocalcin/biosynthesis , Osteocalcin/genetics , Osteogenesis/drug effects , Osteopontin/genetics , Protein Isoforms/physiology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
10.
Eur Rev Med Pharmacol Sci ; 23(3): 949-957, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30779060

ABSTRACT

OBJECTIVE: This study aims to investigate whether circ-VANGL1 can promote the progression of osteoporosis (OP) by absorbing miRNA-217 to regulate RUNX2 expression. PATIENTS AND METHODS: The serum levels of circ-VANGL1, miRNA-217 and RUNX2 in OP patients and non-OP patients were detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). Their expression levels in human bone marrow mesenchymal stem cells (hBMSCs) at different time points of osteogenesis differentiation were determined as well. The expression levels of RUNX2 and osteogenic proteins (BSP, OCN, OPN) in hBMSCs were detected by Western blot. Dual-Luciferase reporter gene assay was performed to verify the relationship among circ-VANGL1, miRNA-217 and RUNX2. Alkaline phosphatase (ALP) staining was conducted to evaluate the degree of osteogenic differentiation influenced by circ-VANGL1 and miRNA-217. RESULTS: OP patients presented a higher serum level of miRNA-217 and lower serum levels of circ-VANGL1 and RUNX2 relative to non-OP patients. Circ-VANGL1 accelerated osteogenic differentiation by absorbing miRNA-217 to regulate RUNX2 expression. Moreover, miRNA-217 inhibited osteogenic differentiation by degrading RUNX2 by targeting to RUNX2 3'UTR. The overexpression of circ-VANGL1 upregulated expressions of RUNX2, BSP, OCN, and OPN. Meanwhile, ALP activity increased in hBMSCs overexpressing circ-VANGL1. However, co-overexpression of circ-VANGL1 and miRNA-217 did not alter RUNX2 expression. ALP activity in hBMSCs co-overexpressing circ-VANGL1 and miRNA-217 slightly increased, but had no difference with controls. CONCLUSIONS: Circ-VANGL1 promotes the development of OP via binding to miRNA-217 to downregulate RUNX2 expression.


Subject(s)
Carrier Proteins/physiology , Core Binding Factor Alpha 1 Subunit/blood , Membrane Proteins/physiology , MicroRNAs/physiology , Osteoporosis/physiopathology , Carrier Proteins/biosynthesis , Carrier Proteins/blood , Case-Control Studies , Cell Differentiation/physiology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/biosynthesis , Disease Progression , Humans , Integrin-Binding Sialoprotein/biosynthesis , Membrane Proteins/biosynthesis , Membrane Proteins/blood , Mesenchymal Stem Cells/metabolism , MicroRNAs/biosynthesis , MicroRNAs/blood , Osteocalcin/biosynthesis , Osteopontin/biosynthesis , Osteoporosis/blood , Time Factors
11.
J Orthop Surg Res ; 14(1): 11, 2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30621711

ABSTRACT

OBJECTIVE: Psoralen is a natural plant toxin which has the function of protecting fungi, insects, and herbivores. In this study, we aim to investigate the effect and mechanism of psoralen on steroid-induced avascular necrosis of femoral head (SANFH). METHODS: Thirty rabbits were randomly divided into blank group (n = 10), model group (n = 10), and experimental group (n = 10). Rabbits in blank and model groups were treated with normal saline, and rabbits in experimental group were treated with psoralen. Total RNA of bone marrow was extracted by trizol, and the mRNA expression of PPARγ and osteocalcin were detected by q-PCR. Then, the mRNA expression of PPARγ and osteocalcin in the three groups were compared. Western blot was used to detect the PPARγ protein expression in the bone of rabbits. ELISA was used to measure the osteocalcin protein. RESULTS: The mRNA expression of PPARγ in model group significantly increased compared with blank group. The mRNA expression of osteocalcin in model group decreased compared with blank group. However, the mRNA and protein expressions of PPARγ in experimental group were significantly decreased compared with the model group. The protein expressions of osteocalcin increased compared with the model group. There was no significant difference of trabecular bone area (TBA) between experimental and blank groups (P > 0.05). TBA in model group was lower than the experimental group (P < 0.05). There was no significant difference of TBA between experimental and blank groups (P > 0.05). CONCLUSION: This research confirms that psoralen plays a positive role in the rehabilitation of SANFH.


Subject(s)
Cancellous Bone/metabolism , Femur Head Necrosis/metabolism , Ficusin/pharmacology , Osteocalcin/biosynthesis , PPAR gamma/biosynthesis , Steroids/toxicity , Animals , Cancellous Bone/drug effects , Femur Head Necrosis/chemically induced , Femur Head Necrosis/drug therapy , Ficusin/therapeutic use , Gene Expression , Osteocalcin/genetics , PPAR gamma/genetics , Rabbits , Random Allocation
12.
J Pharmacol Sci ; 138(3): 209-213, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30389276

ABSTRACT

Inorganic polyphosphate (polyP), a linear polymer of orthophosphate, is found at high concentrations in osteoblasts. We demonstrated the effects of various polyP concentrations on the mineralization of rat osteoblast ROS17/2.8 cells. Mineralization of ROS17/2.8 was induced by a high polyP concentration (1 mg/mL), which was accompanied by an upregulation of the bone sialoprotein and osteocalcin. In contrast, a low polyP concentration (1 × 10-2 mg/mL) reduced mineralization without affecting the osteogenic gene expression. Furthermore, gene expression profiling and forced expression analysis indicated that phosphodiesterase 11a could be a candidate involved in the dose-dependent effect of polyP on osteoblast mineralization.


Subject(s)
Calcification, Physiologic/drug effects , Osteoblasts/metabolism , Polyphosphates/pharmacology , Animals , Calcification, Physiologic/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression/drug effects , Gene Expression Profiling , Osteocalcin/biosynthesis , Osteopontin/biosynthesis , Phosphoric Diester Hydrolases/biosynthesis , Rats
13.
Heart Surg Forum ; 21(5): E375-E381, 2018 Sep 13.
Article in English | MEDLINE | ID: mdl-30311888

ABSTRACT

BACKGROUND: Chemically cross-linked animal tissues, such as porcine aortic valves (PAVs) have many documented advantages over mechanical valves. However, calcification is the major underlying pathologic process that results in bioprosthetic valve failure. Recently, several reports described the expression of noncollagenous bone matrix proteins in bioprosthetic valves and suggested an actively regulated process of tissue repair. METHODS: Thirty-one explanted PAVs with evidence of calcification were collected and examined for the protein expression implicated in myofibroblast activation, osteoblast differentiation, and bone matrix deposition by using immunohistochemistry. RESULTS: The mean duration that PAVs were implanted was 11.5 ± 5.6 years, ranging from 12 months to 28 years. Pearson correlation analysis showed a significant relationship between the duration and valvular calcification (r = 0.3818, P = .034). The number of vimentin-positive mesenchymal cells in explanted PAVs was significantly lower than that of unused PAVs (P < .01). However, increased expression of α-smooth muscle actin (α-SMA) (P < .01), proliferating cell nuclear antigen (PCNA, P < .01), Cbfa1/Runx2 (P < .01), osterix (P = .0126), bone sialoprotein (BSP, P < .01), osteocalcin (P < .01), and osteopontin (P < .01) was found in explanted PAVs. Immunohistochemical staining of alkaline phosphatase (ALP) and osteocalcin was negative in the unused PAVs. In explanted PAVs, the expression level of these 2 proteins was also significantly increased. CONCLUSIONS: Our results support the view that PAV calcification is an actively regulated process with osteogenic signaling activation.


Subject(s)
Actins/biosynthesis , Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Bioprosthesis , Calcinosis/metabolism , Heart Valve Prosthesis , Osteocalcin/biosynthesis , Osteopontin/biosynthesis , Sp7 Transcription Factor/biosynthesis , Adult , Aged , Animals , Aortic Valve/metabolism , Aortic Valve/surgery , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/surgery , Biomarkers/metabolism , Calcinosis/pathology , Calcinosis/surgery , Female , Humans , Immunohistochemistry , Male , Middle Aged , Proliferating Cell Nuclear Antigen/metabolism , Retrospective Studies , Swine , Young Adult
14.
Cytotherapy ; 20(10): 1267-1277, 2018 10.
Article in English | MEDLINE | ID: mdl-30196010

ABSTRACT

BACKGROUND AIMS: Regenerative medicine strategies based on cell therapy are considered a promising approach to repair bone defects. The aims of this study were to evaluate the effect of subculturing on the osteogenic potential of osteoblasts derived from newborn rat calvaria and the effect of these osteoblasts on bone repair of rat calvaria defects. METHODS: Cells were obtained from 50 newborn rat calvaria, and primary osteoblasts (OB) were compared with first passage (OB-P1) in terms of osteogenic potential by assaying cell proliferation, alkaline phosphatase (ALP) activity, extracellular matrix mineralization and gene expression of the osteoblastic markers RUNX2, ALP, osteocalcin and bone sialoprotein. Then, 5-mm calvaria defects were created in 24 Wistar rats, and after 2 weeks, they were locally injected with 50 µL of phosphate-buffered saline containing either 5 × 106 osteoblasts (OB-P1, n = 12) or no cells (control, n = 12). Four weeks post-injection, the bone formation was evaluated by micro-computed tomography and histological analyses. Data were compared by analysis of variance, followed by the Student-Newman-Keuls's test or Student's t-test (P ≤ 0.05). RESULTS: OB-P1 showed high proliferation and ALP activity, and despite the reduced gene expression of osteoblastic markers and extracellular matrix mineralization compared with OB, they displayed osteogenic potential, being a good choice for injection into calvaria defects. The micro-tomographic and histological data showed that defects treated with OB-P1 presented higher bone formation compared with control defects. DISCUSSION: Our results indicate that cells derived from newborn rat calvaria retain osteoblastic characteristics after subculturing and that these osteoblasts stimulate bone repair in a rat calvaria defect model.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Osteoblasts/transplantation , Skull/injuries , Alkaline Phosphatase/metabolism , Animals , Biomarkers , Cells, Cultured , Extracellular Matrix/metabolism , Gene Expression Regulation , Osteoblasts/metabolism , Osteoblasts/physiology , Osteocalcin/biosynthesis , Osteocalcin/genetics , Osteogenesis/physiology , Rats, Wistar , Skull/cytology , Transplantation, Homologous/methods , X-Ray Microtomography
15.
Mol Biol Rep ; 45(5): 1289-1296, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30066297

ABSTRACT

Canine primary bone tumors have a plastic radiographic image, demanding histopathological confirmation. Bone tumors are characterized by the type and amount of extracellular matrix produced what cannot be easily recognized, especially in biopsy samples. Identifying cellular markers that could aid diagnosis has supported various studies in oncological pathology. This study aimed to evaluate 22 canine primary bone neoplasms, establishing their histopathological diagnosis and evaluated vimentin, osteonectin and osteocalcin expression and their implication in diagnosis and prognosis. There were 12 productive osteoblastic osteosarcomas, six minimally productive osteoblastic osteosarcoma, two chondrosarcomas, one fibrosarcoma and one hemangiosarcoma. Immunostaining was cytoplasmatic in all cases, with average percentage of 87.9% for vimentin, 98.0% for osteonectin and 99.9% for osteocalcin. In this last case, only osteosarcomas were considered. Intensity was higher in vimentin labeling (+++), followed by osteonectin (++) and osteocalcin (+). One osteosarcoma showed negative immunostaining for vimentin and of samples submitted to anti-osteocalcin immunostaining, three osteosarcomas and one fibrosarcoma had negative staining. Besides identifying mesenchymal origin, vimentin elevated expression in canine bone tumors can be related to epithelial-mesenchymal transition, leading to more aggressive tumoral phenotypes and metastasis development. Similarly, high osteonectin expression is implicated in neoplastic cell invasion and is also related to metastasis spread. Decreased osteocalcin expression was found in some osteosarcoma samples and can be related to poor prognosis, as in human osteosarcomas. Our findings suggest that vimentin, osteonectin and osteocalcin not only aid diagnosis but can be related to prognosis in canine primary bone tumors, especially osteosarcomas and its osteoblastic subtype.


Subject(s)
Bone Neoplasms/veterinary , Dog Diseases/metabolism , Osteocalcin/biosynthesis , Osteonectin/biosynthesis , Vimentin/biosynthesis , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Diagnosis, Differential , Dog Diseases/diagnosis , Dog Diseases/genetics , Dogs , Immunohistochemistry , Osteocalcin/genetics , Osteocalcin/metabolism , Osteonectin/genetics , Osteonectin/metabolism , Osteosarcoma/diagnosis , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/veterinary , Prognosis , Transcriptome , Vimentin/genetics , Vimentin/metabolism
16.
Int J Mol Sci ; 19(8)2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30071658

ABSTRACT

A chemically-treated titanium alloy (Ti6Al4V) surface, able to induce hydroxyapatite precipitation from body fluids (inorganic mineralization activity), was functionalized with a polyphenolic extract from green tea (tea polyphenols, TPH). Considering that green tea polyphenols have stimulating effects on bone forming cells (biological mineralization), the aim was to test their osteoinductive behavior due to co-operation of inorganic and biological mineralization on mesenchymal stem cells KUSA A1. The functionalized surfaces were characterized by using the Folin⁻Ciocalteu method and X-ray photoelectron spectroscopy to confirm the successful outcome of the functionalization process. Two cell cultures of mesenchymal stem cells, KUSA A1 were performed, with or without osteoinductive factors. The cells and surfaces were characterized for monitoring cell viability and hydroxyapatite production: Fourier Transform Infrared Spectroscopy and Raman spectroscopy analyses showed deposition of hydroxyapatite and collagen due to the cell activity, highlighting differentiation of KUSA A1 into osteoblasts. A higher production of extracellular matrix was highlighted on the functionalized samples by laser microscope and the fluorescence images showed higher viability of cells and greater presence of osteocalcin in these samples. These results highlight the ability of polyphenols to improve cell differentiation and to stimulate biological mineralization, showing that surface functionalization of metal implants could be a promising way to improve osteointegrability.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Polyphenols/pharmacology , Tea/chemistry , Titanium/pharmacology , Alloys , Cell Line , Coated Materials, Biocompatible/chemistry , Durapatite/metabolism , Extracellular Matrix/metabolism , Humans , Mesenchymal Stem Cells/cytology , Osteocalcin/biosynthesis , Polyphenols/chemistry , Titanium/chemistry
17.
Bosn J Basic Med Sci ; 18(4): 328-335, 2018 Nov 07.
Article in English | MEDLINE | ID: mdl-30054999

ABSTRACT

Numerous studies indicated that microRNAs are critical in the regulation of cellular differentiation, by controlling the expression of underlying genes. The aim of this study was to investigate the effect of miR-210 upregulation on differentiation of human umbilical cord blood (HUCB)-derived mesenchymal stem cells (MSCs) into osteoblasts. MSCs were isolated from HUCB and confirmed by their adipogenic/osteogenic differentiation and flow cytometric analysis of surface markers. Pre-miR-210 was amplified from human DNA, digested and ligated with plenti-III-mir-green fluorescent protein (GFP) vector, and cloned in STBL4 bacteria. After confirmation with polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), the plenti-III-GFP segment bearing pre-miR-210 was transfected into MSCs by electroporation. Two control vectors, pmaxGFP and Scramble, were transfected separately into MSCs. The expression of miR-210 and genes related to osteoblast differentiation, i.e., runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and osteocalcin gene, in the three groups of transfected MSCs was analyzed 0, 7, 14, and 21 days of transfection by quantitative reverse transcription PCR (qRT-PCR). Overexpression of miR-210 was observed in MSCs transfected with miR-210-bearing plasmid, and this was significantly different compared to Scramble group (p < 0.05). Significantly increased expression of Runx2 (at day 7 and 14), ALP and osteocalcin genes (at all time points for both genes) was observed in MSCs with miR-210-bearing plasmid compared to controls. Overall, the overexpression of miR-210 in MSCs led to MSC differentiation into osteoblasts, most probably by upregulating the Runx2, ALP, and osteocalcin genes at different stages of cell differentiation. Our study confirms the potential of miRNAs in developing novel therapeutic strategies that could target regulatory mechanisms of cellular differentiation in various disease states.


Subject(s)
Mesenchymal Stem Cells/drug effects , MicroRNAs/biosynthesis , Osteoblasts/drug effects , Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/biosynthesis , Green Fluorescent Proteins , Humans , MicroRNAs/genetics , Osteocalcin/biosynthesis , Osteogenesis , Polymorphism, Restriction Fragment Length , Up-Regulation
18.
Photomed Laser Surg ; 36(8): 406-414, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29851368

ABSTRACT

OBJECTIVE: We examined the effects of photobiomodulation (PBM) on stereological parameters, and gene expression of Runt-related transcription factor 2 (RUNX2), osteocalcin, and receptor activator of nuclear factor kappa-B ligand (RANKL) in repairing tissue of tibial bone defect in streptozotocin (STZ)-induced type 1 diabetes mellitus (TIDM) in rats during catabolic response of fracture healing. BACKGROUND DATA: There were conflicting results regarding the efficacy of PBM on bone healing process in healthy and diabetic animals. MATERIALS AND METHODS: Forty-eight rats have been distributed into four groups: group 1 (healthy control, no TIDM and no PBM), group 2 (healthy test, no TIDM and PBM), group 3 (diabetic control, TIDM and no PBM), and group 4 (diabetic test, no TIDM and PBM). TIDM was induced in the groups 3 and 4. A partial bone defect in tibia was made in all groups. The bone defects of groups second and fourth were irradiated by a laser (890 nm, 80 Hz, 1.5 J/cm2). Thirty days after the surgery, all bone defects were extracted and were submitted to stereological examination and real-time polymerase chain reaction (RT-PCR). RESULTS: PBM significantly increased volumes of total callus, total bone, bone marrow, trabecular bone, and cortical bone, and the numbers of osteocytes and osteoblasts of callus in TIDM rats compared to those of callus in diabetic control. In addition, TIDM increased RUNX2, and osteocalcin in callus of tibial bone defect compared to healthy group. PBM significantly decreased osteocalcin gene expression in TIDM rats. CONCLUSIONS: PBM significantly increased many stereological parameters of bone repair in an STZ-induced TIDM during catabolic response of fracture healing. Further RT-PCR test demonstrated that bone repair was modulated in diabetic rats during catabolic response of fracture healing by significant increase in mRNA expression of RUNX2, and osteocalcin compared to healthy control rats. PBM also decreased osteocalcin mRNA expression in TIDM rats.


Subject(s)
Fracture Healing/radiation effects , Low-Level Light Therapy , Osteotomy , Tibia/radiation effects , Tibial Fractures/radiotherapy , Animals , Core Binding Factor Alpha 1 Subunit/biosynthesis , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/complications , Disease Models, Animal , Female , Fracture Healing/physiology , Osteocalcin/biosynthesis , RANK Ligand/biosynthesis , Rats , Rats, Wistar , Tibia/physiopathology , Tibial Fractures/complications , Tibial Fractures/physiopathology , Tibial Fractures/therapy
19.
J Appl Oral Sci ; 26: e20170367, 2018 Jun 11.
Article in English | MEDLINE | ID: mdl-29898175

ABSTRACT

OBJECTIVES: To study the intensity of inflammatory infiltrate and production of interleukin-1ß (ll-1ß), tumor necrosis factor-ß (TNF-ß), fibroblast growth factor-2 (FGF-2), glutathione peroxidase (GPX), and osteocalcin in response to in-office tooth bleaching in rats. MATERIAL AND METHODS: Twenty male Wistar rats were randomized into four groups (n=5) according to the received treatment (tooth bleaching or no treatment - control) and the period of euthanasia after treatment (24 h or 10 days). We performed tooth bleaching using a 38% hydrogen peroxide gel on maxillary and mandibular incisors. After euthanasia, incisors (20 per group) were processed for histological analysis, immunohistochemistry staining of ll-1ß, TNF-ß, FGF-2 and GPX and osteocalcin by immunofluorescence. We analyzed data using the Mann-Whitney and Kruskal-Wallis/Dunn tests (p<0.05). RESULTS: The bleached groups presented statistically significant differences regarding the pulp inflammation stage compared with the control groups. Bleached teeth showed moderate/severe inflammatory infiltrate and control groups presented absent inflammatory cells or a negligible number of mononuclear cells (p<0.001) at two times (24 h and 10 days). There was strong staining for ll-1ß, TNF-ß, and GPX in bleached groups at 24 h and strong staining for ll-1ß, TNF-ß, GPX and FGF-2 at 10 days. After 10 days of tooth bleaching, the bleached group showed a statistically superior amount of osteocalcin than the other groups (p<0.01). CONCLUSIONS: Tooth bleaching with 38% hydrogen peroxide causes severe pulp inflammation, but characteristics of tissue repair after 10 days.


Subject(s)
Hydrogen Peroxide/adverse effects , Pulpitis/chemically induced , Pulpitis/pathology , Tooth Bleaching Agents/administration & dosage , Tooth Bleaching/adverse effects , Animals , Fibroblast Growth Factor 2/biosynthesis , Glutathione Peroxidase/biosynthesis , Immunohistochemistry , Interleukin-1beta/biosynthesis , Lymphotoxin-alpha/biosynthesis , Male , Microscopy, Fluorescence , Osteocalcin/biosynthesis , Pulpitis/metabolism , Random Allocation , Rats, Wistar , Time Factors
20.
J Cell Physiol ; 233(3): 2213-2224, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28688217

ABSTRACT

Yes-associated protein 1 (YAP1) transcriptional coactivator is a mediator of mechanosensitive signaling. Cementum, which covers the tooth root surface, continuously senses external mechanical stimulation. Cementoblasts are responsible for the mineralization and maturation of the cementum. However, the effect of YAP1 on cementoblast differentiation remains largely unknown. In this study, we initially demonstrated that YAP1 overexpression enhanced the mineralization ability of cementoblasts. YAP1 upregulated the mRNA and protein expression of several cementogenesis markers, such as alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and dentin matrix acidic phosphoprotein 1 (DMP1). The YAP1 overexpression group showed higher intensities of ALP and Alizarin red stain than the YAP1-knockdown group. Unexpectedly, a sharp increase in the expression of dentin sialophosphoprotein (DSPP) was induced by the overexpression of YAP1. Knockdown of YAP1 suppressed DSPP transcriptional activity. YAP1 overexpression activated Smad-dependent BMP signaling and slightly inhibited Erk1/2 signaling pathway activity. Treatment with specific BMP antagonist (LDN193189) prevented the upregulation of the mRNA levels of ALP, RUNX2, and OCN, as well as intensity of ALP-stained and mineralized nodules in cementoblasts. The Erk1/2 signaling pathway inhibitor (PD 98,059) upregulated these cementogenesis markers. Thus, our study suggested that YAP1 enhanced cementoblast mineralization in vitro. YAP1 exerted its effect on the cementoblast partly by regulating the Smad-dependent BMP and Erk1/2 signaling pathways.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Morphogenetic Protein 1/metabolism , Cementogenesis/physiology , Dental Cementum/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphoproteins/metabolism , Smad Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Alkaline Phosphatase/biosynthesis , Animals , Bone Morphogenetic Protein 1/antagonists & inhibitors , Cell Cycle Proteins , Cell Differentiation , Cell Line , Core Binding Factor Alpha 1 Subunit/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Mice , Osteocalcin/biosynthesis , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sialoglycoproteins/biosynthesis , Sialoglycoproteins/genetics , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...