Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biotechnol Bioeng ; 120(10): 3067-3078, 2023 10.
Article in English | MEDLINE | ID: mdl-37317560

ABSTRACT

Adipose tissue is an attractive source of mesenchymal stem cells (at-MSCs), but their low osteogenic potential limits their use in bone regeneration. Adipose tissue plays a role in pro-inflammatory diseases by releasing cytokines with a catabolic effect on bone, such as tumor necrosis factor-alpha (TNF-α). Thus, we hypothesized that endogenous TNF-α could have a negative effect on at-MSC differentiation into osteoblasts. Short interfering RNAs (siRNAs) targeting TNF-α receptors (siR1, siR2, and si1R/R2) were transfected into at-MSCs, and cell differentiation was assessed by measuring the expression of bone markers, ALP activity, and mineralized matrix. Scrambled was used as Control. Knockout at-MSCs (KOR1/R2) was injected in mice calvaria defects, and bone formation was evaluated by microtomography and histological analysis. Data were compared by Kruskal-Wallis or analysis of variance (5%). The expression of bone markers confirmed that at-MSCs differentiate less than bone marrow MSCs. In silenced cells, the expression of Alp, Runx2, and Opn was generally higher compared to Control. ALP, RUNX2, and OPN were expressed at elevated levels in silenced groups, most notably at-MSCs-siR1/R2. ALP was detected at high levels in at-MSCs-siR1/R2 and in-MSCs-siR1, followed by an increase in mineralized nodules in at-MSCs-siR1/R2. As the morphometric parameters increased, the groups treated with KOR1/R2 exhibited slight bone formation near the edges of the defects. Endogenous TNF-α inhibits osteoblast differentiation and activity in at-MSCs, and its disruption increases bone formation. While opening a path of investigation, that may lead to the development of new treatments for bone regeneration using at-MSC-based therapies.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Tumor Necrosis Factor-alpha , Animals , Mice , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Mice, Knockout , Osteoblasts , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Front Oral Health ; 1: 5, 2020.
Article in English | MEDLINE | ID: mdl-35047978
3.
J Biol Chem ; 293(45): 17646-17660, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30242124

ABSTRACT

MicroRNAs (miRs) and Hox transcription factors have decisive roles in postnatal bone formation and homeostasis. In silico analysis identified extensive interaction between HOXA cluster mRNA and microRNAs from the miR-23a cluster. However, Hox regulation by the miR-23a cluster during osteoblast differentiation remains undefined. We examined this regulation in preosteoblasts and in a novel miR-23a cluster knockdown mouse model. Overexpression and knockdown of the miR-23a cluster in preosteoblasts decreased and increased, respectively, the expression of the proteins HOXA5, HOXA10, and HOXA11; these proteins' mRNAs exhibited significant binding with the miR-23a cluster miRNAs, and miRNA 3'-UTR reporter assays confirmed repression. Importantly, during periods correlating with development and differentiation of bone cells, we found an inverse pattern of expression between HoxA factors and members of the miR-23a cluster. HOXA5 and HOXA11 bound to bone-specific promoters, physically interacted with transcription factor RUNX2, and regulated bone-specific genes. Depletion of HOXA5 or HOXA11 in preosteoblasts also decreased cellular differentiation. Additionally, stable overexpression of the miR-23a cluster in osteoblasts decreased the recruitment of HOXA5 and HOXA11 to osteoblast gene promoters, significantly inhibiting histone H3 acetylation. Heterozygous miR-23a cluster knockdown female mice (miR-23a ClWT/ZIP) had significantly increased trabecular bone mass when compared with WT mice. Furthermore, miR-23a cluster knockdown in calvarial osteoblasts of these mice increased the recruitment of HOXA5 and HOXA11, with a substantial enrichment of promoter histone H3 acetylation. Taken together, these findings demonstrate that the miR-23a cluster is required for maintaining stage-specific HoxA factor expression during osteogenesis.


Subject(s)
3' Untranslated Regions , Cell Differentiation , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Multigene Family , Osteoblasts/metabolism , Phosphoproteins/metabolism , Acetylation , Animals , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , HEK293 Cells , Histones/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Humans , Mice , MicroRNAs/genetics , Osteoblasts/cytology , Osteogenesis , Phosphoproteins/genetics , Transcription Factors
4.
J Biomed Mater Res A ; 105(10): 2783-2788, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28643442

ABSTRACT

Several studies have shown the positive effects of Ti either with nanotopography or coated with collagen on osteoblast differentiation. Thus, we hypothesized that the association of nanotopography with collagen may increase the in vitro osteogenesis on Ti surface. Ti discs with nanotopography with or without collagen coating were characterized by scanning electron microscopy and atomic force microscopy. Rat calvaria-derived osteoblastic cells were cultured on both Ti surfaces for up to 14 days and the following parameters were evaluated: cell proliferation, alkaline phosphatase (ALP) activity, extracellular matrix mineralization, protein expression of bone sialoprotein (BSP) and osteopontin (OPN), and gene expression of collagen type 1a (Coll1a), runt-related transcription factor 2 (Runx2), osterix (OSX), osteocalcin (OC), Ki67, Survivin, and Bcl2-associated X protein (BAX). Surface characterization evidenced that collagen coating did not alter the nanotopography. Collagen coating increased cell proliferation, ALP activity, extracellular matrix mineralization, and Coll1a, OSX, OC, and BAX gene expression. Also, OPN and BSP proteins were strongly detected in cultures grown on both Ti surfaces. In conclusion, our results showed that the combination of nanotopography with collagen coating stimulates the early, intermediate, and final events of the in vitro osteogenesis and may be considered a potential approach to promote osseointegration of Ti implants. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2783-2788, 2017.


Subject(s)
Coated Materials, Biocompatible/chemistry , Collagen/chemistry , Nanostructures/chemistry , Osteoblasts/cytology , Osteogenesis , Titanium/chemistry , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Osteoblasts/metabolism , Rats , Rats, Wistar , Surface Properties
5.
J Biomed Mater Res A ; 105(2): 419-423, 2017 02.
Article in English | MEDLINE | ID: mdl-27682446

ABSTRACT

The ability of Biosilicate® with two crystalline phases (BioS-2P) to drive osteoblast differentiation encourages the investigation of the cellular mechanisms involved in this process. Then, the aim of our study was to analyze the large-scale gene expression of osteoblasts grown on BioS-2P compared with Bioglass® 45S5 (45S5). Osteoblasts differentiated from rat bone marrow mesenchymal stem cells were cultured under osteogenic conditions on BioS-2P, 45S5 and polystyrene (control). After 10 days, the expression of 23,794 genes was analyzed using mRNA Sequencing and the data were validated by real-time PCR. The BioS-2P exhibited 5 genes upregulated and 3 downregulated compared with 45S5. Compared with control, BioS-2P upregulated 15 and downregulated 11 genes, while 45S5 upregulated 25 and downregulated 21 genes. Eight genes were commonly upregulated and 4 downregulated by both bioactive glasses. In conclusion, our results demonstrated that bioactive glasses affect the gene expression profiling of osteoblasts. Most of the regulated genes by both BioS-2P and 45S5 are associated with the process of mineralization highlighting their osteostimulation property that is, at least in part, derived from the ability to modulate the intracellular machinery to promote osteoblast genotype expression. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 419-423, 2017.


Subject(s)
Calcification, Physiologic/drug effects , Ceramics/pharmacology , Gene Expression Regulation/drug effects , Osteoblasts/metabolism , Animals , Gene Expression Profiling , Male , Osteoblasts/cytology , Rats , Rats, Wistar , Surface Properties
6.
J Biomater Sci Polym Ed ; 27(13): 1369-79, 2016 09.
Article in English | MEDLINE | ID: mdl-27312544

ABSTRACT

The poly(vinylidene-trifluoroethylene)/barium titanate (PVDF) membrane enhances in vitro osteoblast differentiation and in vivo bone repair. Here, we hypothesized that this higher bone repair could be also due to bone resorption inhibition mediated by a microRNA (miR)/RANKL circuit. To test our hypothesis, the large-scale miR expression of bone tissue grown on PVDF and polytetrafluoroethylene (PTFE) membranes was evaluated to identify potential RANKL-targeted miRs modulated by PVDF. The animal model used was rat calvarial defects implanted with either PVDF or PTFE. At 4 and 8 weeks, the bone tissue grown on membranes was submitted to a large-scale analysis of miRs by microarray. The expression of miR-34a and some of its targets, including RANKL, were evaluated by real-time polimerase chain reaction and osteoclast activity was detected by tartrate-resistant acid phosphatase (TRAP) staining. Among more than 250 miRs, twelve, including miR-34a, were simultaneously higher expressed (≥2 fold) at 4 and 8 weeks on PVDF. The higher expression of miR-34a was concomitant with a reduced expression of all its evaluated targets, including RANKL. Additionally, more TRAP-positive cells were observed in bone tissue grown on PTFE compared with PVDF in both time points. In conclusion, our results suggest that the higher bone formation induced by PVDF could be, at least in part, triggered by a miR-34a increase and RANKL decrease, which may inhibit osteoclast differentiation and activity, and bone resorption.


Subject(s)
Barium Compounds/chemistry , Bone Regeneration , Hydrocarbons, Fluorinated/chemistry , MicroRNAs/metabolism , Osteoblasts/cytology , RANK Ligand/metabolism , Titanium/chemistry , Vinyl Compounds/chemistry , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone and Bones/metabolism , Cell Differentiation , Gene Expression , Membranes, Artificial , Osteoblasts/metabolism , Osteogenesis , Rats, Wistar
7.
Mol Cell Biol ; 35(18): 3116-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26124283

ABSTRACT

Studies of proteins involved in microRNA (miRNA) processing, maturation, and silencing have indicated the importance of miRNAs in skeletogenesis, but the specific miRNAs involved in this process are incompletely defined. Here, we identified miRNA 665 (miR-665) as a potential repressor of odontoblast maturation. Studies with cultured cell lines and primary embryonic cells showed that miR-665 represses the expression of early and late odontoblast marker genes and stage-specific proteases involved in dentin maturation. Notably, miR-665 directly targeted Dlx3 mRNA and decreased Dlx3 expression. Furthermore, RNA-induced silencing complex (RISC) immunoprecipitation and biotin-labeled miR-665 pulldown studies identified Kat6a as another potential target of miR-665. KAT6A interacted physically and functionally with RUNX2, activating tissue-specific promoter activity and prompting odontoblast differentiation. Overexpression of miR-665 reduced the recruitment of KAT6A to Dspp and Dmp1 promoters and prevented KAT6A-induced chromatin remodeling, repressing gene transcription. Taken together, our results provide novel molecular evidence that miR-665 functions in an miRNA-epigenetic regulatory network to control dentinogenesis.


Subject(s)
Dentin/embryology , Dentinogenesis/genetics , Gene Expression Regulation, Developmental/genetics , MicroRNAs/genetics , Odontoblasts/cytology , Animals , Cell Differentiation/genetics , Cell Line , Chromatin/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Silencing , HEK293 Cells , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Homeodomain Proteins/genetics , Humans , Mice , MicroRNAs/biosynthesis , Peptide Hydrolases/biosynthesis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic/genetics , Rats , Sialoglycoproteins/metabolism , Transcription Factors/genetics
8.
J Cell Physiol ; 229(11): 1690-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24619927

ABSTRACT

The aim of this study was to investigate if chemically produced nanotopography on titanium (Ti) surface induces osteoblast differentiation of cultured human bone marrow mesenchymal stem cells (hMSCs) by regulating the expression of microRNAs (miRs). It was demonstrated that Ti with nanotopography induces osteoblast differentiation of hMSCs as evidenced by upregulation of osteoblast specific markers compared with untreated (control) Ti at day 4. At this time-point, miR-sequencing analysis revealed that 20 miRs were upregulated (>twofold) while 20 miRs were downregulated (>threefold) in hMSCs grown on Ti with nanotopography compared with control Ti. Three miRs, namely miR-4448, -4708, and -4773, which were significantly downregulated (>fivefold) by Ti with nanotopography affect osteoblast differentiation of hMSCs. These miRs directly target SMAD1 and SMAD4, both key transducers of the bone morphogenetic protein 2 (BMP-2) osteogenic signal, which were upregulated by Ti with nanotopography. Overexpression of miR-4448, -4708, and 4773 in MC3T3-E1 pre-osteoblasts noticeably inhibited gene and protein expression of SMAD1 and SMAD4 and therefore repressed the gene expression of key bone markers. Additionally, it was observed that the treatment with BMP-2 displayed a higher osteogenic effect on MC3T3-E1 cells grown on Ti with nanotopography compared with control Ti, suggesting that the BMP-2 signaling pathway was more effective on this surface. Taken together, these results indicate that a complex regulatory network involving a miR-SMAD-BMP-2 circuit governs the osteoblast differentiation induced by Ti with nanotopography. J. Cell. Physiol. 229: 1690-1696, 2014. © 2014 Wiley Periodicals, Inc.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Cell Lineage , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Nanoparticles/chemistry , Osteoblasts/cytology , Smad Proteins/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Lineage/drug effects , Cell Lineage/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Mice , MicroRNAs/metabolism , Middle Aged , Osteocalcin/metabolism , Osteopontin/metabolism , Titanium/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
9.
J Biomed Mater Res A ; 102(4): 991-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23625878

ABSTRACT

In this study, we evaluated the effect of new plasma-nitrided Ti surfaces on the progression of osteoblast cultures, including cell adhesion, proliferation and differentiation. Ti surfaces were treated using two plasma-nitriding protocols, hollow cathode for 3 h (HC 3 h) and 1 h (HC 1 h) and planar for 1 h. Untreated Ti surfaces were used as control. Cells derived from human alveolar and rat calvarial bones were cultured on Ti surfaces for periods of up to 14 days and the following parameters were evaluated: cell morphology, adhesion, spreading and proliferation, alkaline phosphatase (ALP) activity, extracellular matrix mineralization, and gene expression of key osteoblast markers. Plasma-nitriding treatments resulted in Ti surfaces with distinct physicochemical characteristics. The cell adhesion and ALP activity were higher on plasma-nitrided Ti surfaces compared with untreated one, whereas cell proliferation and extracellular matrix mineralization were not affected by the treatments. In addition, the plasma-nitrided Ti surfaces increased the ALP, reduced the osteocalcin and did not affect the Runx2 gene expression. We have shown that HC 3 h and planar Ti surfaces slightly favored the osteoblast differentiation process, and then these surfaces should be considered for further investigation using preclinical models.


Subject(s)
Cell Differentiation/drug effects , Osteoblasts/cytology , Plasma Gases/pharmacology , Titanium/pharmacology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation/drug effects , Humans , Microscopy, Atomic Force , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Rats , Rats, Wistar , Surface Properties , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...