Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
BMC Oral Health ; 24(1): 390, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38539170

RESUMO

BACKGROUND: Periodontal ligament stem cells (PDLSCs) have been proposed as therapeutic candidates in periodontal diseases and periodontium defects. Paracrine factors of PDLSCs, namely, secretome, can contribute to tissue regeneration comparable to direct stem cell application. This study explored restoration effects of PDLSC-derived secretome/conditioned medium (PDLSC-CM) on PDLSCs themselves in an inflammatory microenvironment and identified its action mechanisms using proteomics and transcriptomic profiling. METHODS: PDLSC-CM was prepared from cells under healthy culture conditions. Mass spectrometry and liquid chromatography-tandem mass spectrometry (LC-MS/MS) were then performed to analyze the PDLSC-CM proteome. Osteogenic differentiation of PDLSCs under inflammatory conditions or in the presence of PDLSC-CM was then characterized in assays of alkaline phosphatase activity, intracellular calcium levels, protein expression of osteogenic markers, and matrix mineralization. Furthermore, the transcriptomic profile was assessed to identify significantly enriched signaling pathways and associated molecular networks by RNA sequencing. RESULTS: LC-MS/MS proteomics identified a total of 203 proteins and distinguished 187 significant protein changes in PDLSC-CM compared to control-CM. LPS-treated PDLSCs significantly attenuated osteogenic differentiation. When PDLSCs were treated with PDLSC-CM alone, their osteogenic activity was significantly upregulated compared to the control group. Moreover, the LPS-impaired osteogenesis of PDLSCs was reconstituted by PDLSC-CM treatment. RNA sequencing revealed 252, 1,326, and 776 differentially expressed genes in the control vs. LPS, control vs. PDLSC-CM, and LPS vs. LPS + PDLSC-CM groups, respectively. CONCLUSION: This study suggest that PDLSC-CM restores the osteogenic potential of PDLSCs in an inflammatory environment through secretory functions representing potential repair and regenerative mechanisms.


Assuntos
Ligamento Periodontal , Periodontite , Humanos , Osteogênese/genética , Meios de Cultivo Condicionados/farmacologia , Proteoma/farmacologia , Transcriptoma , Lipopolissacarídeos/farmacologia , Cromatografia Líquida , Secretoma , Espectrometria de Massas em Tandem , Células-Tronco , Diferenciação Celular , Células Cultivadas
2.
Oral Dis ; 28(6): 1628-1639, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33682270

RESUMO

BACKGROUND/OBJECTIVES: Fucoidan has been focused as a multifunctional therapeutic uses including bone health supplements. However, the critical molecular mechanisms of fucoidan for bone therapeutic agents have not been fully understood. We investigated the osteoinductive effect of fucoidan on periodontal ligament stem cells (PDLSCs) and how this polymer encouraged PDLSC osteogenesis. MATERIALS AND METHODS: Osteogenic induction of PDLSCs was processed by culturing cells with fucoidan treatment. Osteogenic differentiation of PDLSCs was verified by alkaline phosphatase (ALP) activity, matrix mineralization assay, intracellular calcium levels, and mRNA expression and protein levels of osteogenic markers. RESULTS: Fucoidan treatment showed higher osteogenic activity in the PDLSCs than the control groups. PDLSCs with fucoidan also presented increased levels of the phosphatidylinositol-3-kinase (PI3K) isoforms, p110α and p110γ compared to control cells. The phosphorylation of Akt, a PI3K downstream effector, was significantly increased at 90 min of fucoidan induction. Expression of ß-catenin, a coactivator of canonical Wnt pathways, was increased in PDLSCs with fucoidan. ß-catenin was found to link with PI3K activation during the fucoidan stimulation. When cells were blocked by PI3K inhibitor or ß-catenin-specific siRNA, fucoidan-induced osteogenic activity of PDLSCs was significantly attenuated. CONCLUSION: These findings suggest that the fucoidan stimulates osteogenic differentiation of PDLSCs via the PI3K/Akt and Wnt/ß-catenin pathways.


Assuntos
Ligamento Periodontal , Via de Sinalização Wnt , Diferenciação Celular , Células Cultivadas , Osteogênese , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinase/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/farmacologia , Polissacarídeos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células-Tronco/fisiologia , beta Catenina/metabolismo
3.
Mar Drugs ; 20(3)2022 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-35323480

RESUMO

Fucoidan, a marine-sulfated polysaccharide derived from brown algae, has been recently spotlighted as a natural biomaterial for use in bone formation and regeneration. Current research explores the osteoinductive and osteoconductive properties of fucoidan-based composites for bone tissue engineering applications. The utility of fucoidan in a bone tissue regeneration environment necessitates a better understanding of how fucoidan regulates osteogenic processes at the molecular level. Therefore, this study designed a fucoidan and polydopamine (PDA) composite-based film for use in a culture platform for periodontal ligament stem cells (PDLSCs) and explored the prominent molecular pathways induced during osteogenic differentiation of PDLSCs through transcriptome profiling. Characterization of the fucoidan/PDA-coated culture polystyrene surface was assessed by scanning electron microscopy and X-ray photoelectron spectroscopy. The osteogenic differentiation of the PDLSCs cultured on the fucoidan/PDA composite was examined through alkaline phosphatase activity, intracellular calcium levels, matrix mineralization assay, and analysis of the mRNA and protein expression of osteogenic markers. RNA sequencing was performed to identify significantly enriched and associated molecular networks. The culture of PDLSCs on the fucoidan/PDA composite demonstrated higher osteogenic potency than that on the control surface. Differentially expressed genes (DEGs) (n = 348) were identified during fucoidan/PDA-induced osteogenic differentiation by RNA sequencing. The signaling pathways enriched in the DEGs include regulation of the actin cytoskeleton and Ras-related protein 1 and phosphatidylinositol signaling. These pathways represent cell adhesion and cytoskeleton organization functions that are significantly involved in the osteogenic process. These results suggest that a fucoidan/PDA composite promotes the osteogenic potential of PDLSCs by activation of critical molecular pathways.


Assuntos
Hidrogéis/farmacologia , Indóis/farmacologia , Osteogênese/efeitos dos fármacos , Ligamento Periodontal/citologia , Polímeros/farmacologia , Polissacarídeos/farmacologia , Células-Tronco/efeitos dos fármacos , Citoesqueleto de Actina/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidrogéis/química , Indóis/química , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/genética , Polímeros/química , Polissacarídeos/química , Mapas de Interação de Proteínas , Células-Tronco/citologia , Células-Tronco/metabolismo , Propriedades de Superfície , Undaria/química
4.
Int J Mol Sci ; 22(14)2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-34298943

RESUMO

Graphene oxide (GO) is a biocompatible material considered a favorable stem cell culture substrate. In this study, GO was modified with polydopamine (PDA) to facilitate depositing GO onto a tissue culture polystyrene (PT) surface, and the osteogenic performance of the PDA/GO composite in pluripotent embryonic stem cells (ESCs) was investigated. The surface chemistry of the PDA/GO-coated PT surface was analyzed by scanning electron microscopy (SEM) and X-ray photoelectron spectroscopy (XPS). A high cell viability of ESCs cultured on the PDA/GO composite-coated surface was initially ensured. Then, the osteogenic differentiation of the ESCs in response to the PDA/GO substrate was assessed by alkaline phosphatase (ALP) activity, intracellular calcium levels, matrix mineralization assay, and evaluation of the mRNA and protein levels of osteogenic factors. The culture of ESCs on the PDA/GO substrate presented higher osteogenic potency than that on the uncoated control surface. ESCs cultured on the PDA/GO substrate expressed significantly higher levels of integrin α5 and ß1, as well as bone morphogenetic protein receptor (BMPR) types I and II, compared with the control groups. The phosphorylation of extracellular signal-regulated kinase (ERK)1/2, p38, and c-Jun-N-terminal kinase (JNK) mitogen-activated protein kinases (MAPKs) was observed in ESCs culture on the PDA/GO substrate. Moreover, BMP signal transduction by SMAD1/5/8 phosphorylation was increased more in cells on PDA/GO than in the control. The nuclear translocation of SMAD1/5/8 in cells was also processed in response to the PDA/GO substrate. Blocking activation of the integrin α5/ß1, MAPK, or SMAD signaling pathways downregulated the PDA/GO-induced osteogenic differentiation of ESCs. These results suggest that the PDA/GO composite stimulates the osteogenic differentiation of ESCs via the integrin α5/ß1, MAPK, and BMPR/SMAD signaling pathways.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Grafite/farmacologia , Indóis/farmacologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Polímeros/farmacologia , Animais , Técnicas de Cultura de Células , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Cell Physiol Biochem ; 51(2): 746-762, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30463066

RESUMO

BACKGROUND/AIMS: This study investigated the effect of inducible nitric oxide synthase-loaded mineralized nanoparticles (iNOS-MNPs) on the osteogenic differentiation of mouse embryonic stem cells (ESCs). METHODS: We prepared iNOS-MNPs using an anionic block copolymer template-mediated calcium carbonate (CaCO3) mineralization process in the presence of iNOS. iNOS-MNPs were spherical and had a narrow size distribution. iNOS was stably loaded within MNPs without denaturation. In order to confirm the successful introduction of iNOS-MNPs into the cytosol of ESCs, intracellular levels of nitric oxide (NO) was determined with a fluorometric analysis. A NO effector molecule, cyclic guanosine 3',5' monophosphate (cGMP) was also quantified with a competitive enzyme immunoassay. Cell viability in response to iNOS-MNP treatment was determined using the cell counting kit-8 (CCK-8) assay. Alkaline phosphatase (ALP) activity assay, intracellular calcium quantification assay, and Alizarin red S staining for matrix mineralization were performed to investigate osteogenic differentiation of ESCs. The protein levels of Runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), and osterix (OSX) as osteogenic-related factors were also assessed by immunofluorescence staining and Western blot analysis. The complex pathways associated with iNOS-MNP-derived osteogenic differentiation of ESCs were evaluated by network-based analysis. RESULTS: Cells with iNOS-MNPs displayed a significant increase in NO and cGMP concentration compared with the control group. When cells were exposed to iNOS-MNPs, there were no adverse effects on cell viability. Importantly, iNOS-MNP uptake promoted the osteogenic differentiation of ESCs. Using transcriptome profiling, we obtained 1,836 differentially-induced genes and performed functional enrichment analysis with ClueGO and KEGG. These analyses identified significantly enriched and interconnected molecular pathways such as protein kinase activity, estrogen receptor activity, bone morphogenetic protein (BMP) receptor binding, ligand-gated ion channel activity, and phosphatidylinositol 3-phosphate binding. CONCLUSION: These findings suggest that iNOS-MNPs can induce osteogenic differentiation in ESCs by integrating complex signaling pathways.


Assuntos
Diferenciação Celular , Nanopartículas/química , Óxido Nítrico Sintase Tipo II/química , Osteogênese , Fosfatase Alcalina/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , GMP Cíclico/metabolismo , Enzimas Imobilizadas/química , Enzimas Imobilizadas/farmacologia , Redes Reguladoras de Genes , Cinética , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
6.
Biochim Biophys Acta ; 1853(3): 561-72, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25523141

RESUMO

Oxysterols, oxidized derivatives of cholesterol, are biologically active molecules. Specific oxysterols have potent osteogenic properties that act on osteoprogenitor cells. However, the molecular mechanisms underlying these osteoinductive effects on embryonic stem cells (ESCs) are unknown. This study investigated the effect of an oxysterol combination of 22(S)-hydroxycholesterol and 20(S)-hydroxycholesterol (SS) on osteogenic differentiation of ESCs and the alterations to mitochondrial activity during differentiation. Osteogenic differentiation was assessed by alkaline phosphatase (ALP) activity, matrix mineralization, mRNA expression of osteogenic factors, runt-related transcription factor 2, osterix, and osteocalcin, and protein levels of collagen type IA (COLIA) and osteopontin (OPN). Treatment of cells with SS increased osteoinductive activity compared to the control group. Intracellular reactive oxygen species production, intracellular ATP content, mitochondrial membrane potential, mitochondrial mass, mitochondrial DNA copy number, and mRNA expression of peroxisome proliferator-activated receptor-γ coactivators 1α and ß, transcription factors involved in mitochondrial biogenesis, were significantly increased during osteogenesis, indicating upregulation of mitochondrial activity. Oxysterol combinations also increased protein levels of mitochondrial respiratory complexes I-V. We also found that SS treatment increased hedgehog signaling target genes, Smo and Gli1 expression. Inhibition of Hh signaling by cyclopamine suppressed mitochondrial biogenesis and ESC osteogenesis. Subsequently, oxysterol-induced Wnt/ß-catenin pathways were inhibited by repression of Hh signaling and mitochondrial biogenesis. Transfection of ß-catenin specific siRNA decreased the protein levels of COLIA and OPN, as well as ALP activity. Collectively, these data suggest that lipid-based oxysterols enhance differentiation of ESCs toward the osteogenic lineage by regulating mitochondrial activity, canonical Hh/Gli, and Wnt/ß-catenin signaling.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Hidroxicolesteróis/farmacologia , Mitocôndrias/fisiologia , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Transporte de Elétrons/efeitos dos fármacos , Transporte de Elétrons/genética , Células-Tronco Embrionárias/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Camundongos , Osteoblastos/fisiologia , Osteogênese/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
7.
Mol Cell Biochem ; 411(1-2): 83-94, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26369531

RESUMO

Human periodontal ligament fibroblasts (hPLFs) are exposed to oxidative stress during periodontal inflammation and dental treatments. It is hypothesized that hydrogen peroxide (H2O2)-mediated oxidative stress decreases survival and osteogenic differentiation of hPLFs, whereas these decreases are prevented by activation of the Wnt pathway. However, there has been a lack of reports that define the exact roles of canonical Wnt/ß-catenin signaling in H2O2-exposed hPLFs. Treatment with H2O2 reduced viability and proliferation in hPLFs in a dose- and time-dependent manner and led to mitochondria-mediated apoptosis. Pretreatment with lithium chloride (LiCl) or Wnt1 inhibited the oxidative damage that occurred in H2O2-exposed hPLFs. However, knockout of ß-catenin or treatment with DKK1 facilitated the H2O2-induced decreases in viability, mitochondrial membrane potential, and Bcl-2 induction. Osteoblastic differentiation of hPLFs was also inhibited by combined treatment with 100 µM H2O2, as evidenced by the decreases in alkaline phosphatase (ALP) activity and mineralization. H2O2-mediated inhibition of osteoblast differentiation in hPLFs was significantly attenuated in the presence of 500 ng/ml Wnt1 or 20 mM LiCl. In particular, H2O2 stimulated the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) at protein and mRNA levels in hPLFs, whereas the induction was almost completely suppressed in the presence of Wnt1 or LiCl. Furthermore, siRNA-mediated silencing of Nrf2 blocked H2O2-induced decreases in ALP activity and mineralization of hPLFs with the concomitant restoration of runt-related transcription factor 2 and osteocalcin mRNA expression and ALP activity. Collectively, these results suggest that activation of the Wnt/ß-catenin pathway improves proliferation and mineralization in H2O2-exposed hPLFs by downregulating Nrf2.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Ligamento Periodontal/efeitos dos fármacos , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Adulto , Fosfatase Alcalina/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Masculino , Ligamento Periodontal/citologia , Ligamento Periodontal/enzimologia , Adulto Jovem , beta Catenina/genética
8.
Mol Cell Biochem ; 402(1-2): 213-23, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25618247

RESUMO

Canonical Wnt signaling is thought to enhance osteogenic differentiation of human periodontal ligament fibroblasts (hPLFs). However, the mechanism of this enhancement has not yet been defined. We investigated the effects of Wnt1 on osteoblast differentiation of hPLFs and explored the mechanisms of the effects. Treating hPLFs with Wnt1 induced cytosolic accumulation and nuclear translocation of ß-catenin with concomitant increases in alkaline phosphatase (ALP) activity and calcium content in a time-dependent and dose-dependent manner. Wnt1-stimulated differentiation of hPLFs was accompanied by augmented phosphorylation of glycogen synthase kinase (GSK)-3ß and expression of the bone-specific factors runt-related transcription factor 2 (Runx2), osterix2 (Osx2), ALP, type I collagen, osteopontin, and osteocalcin. Pretreatment with Dickkopf-1 inhibited Wnt1-stimulated differentiation of hPLFs by suppressing GSK-3ß phosphorylation, nuclear translocation of ß-catenin, and expression of the bone-specific factors. Small interfering (si) RNA-mediated knockdown of ß-catenin, or pretreatment with FH535, markedly prevented Wnt1-stimulated differentiation of cells by blocking Runx2 and its downstream factors at the mRNA and protein levels. siRNA-mediated silencing of Runx2 also inhibited Wnt1-stimulated mineralization of cells, accompanied by a reduction in the levels of Osx2 and other early and late bone-formation regulatory factors. However, Wnt1-mediated nuclear translocation of ß-catenin and GSK-3ß phosphorylation were not inhibited by knockdown of Runx2 or FH535. Collectively, our findings suggested that Wnt1 stimulates osteogenic differentiation and mineralization of hPLFs, mainly by activating the canonical Wnt/ß-catenin pathway, in which Runx2 is a key downstream regulator.


Assuntos
Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Fibroblastos/fisiologia , Osteoblastos/fisiologia , Ligamento Periodontal/citologia , Proteína Wnt1/fisiologia , Adulto , Fosfatase Alcalina/metabolismo , Calcificação Fisiológica , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Via de Sinalização Wnt , Adulto Jovem , beta Catenina/genética , beta Catenina/metabolismo
9.
Lasers Med Sci ; 30(2): 719-26, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23929563

RESUMO

In our previous studies, we confirmed that low-level laser therapy (LLLT) with a 980-nm gallium-aluminum-arsenide diode laser was beneficial for the healing of the alveolar bone in rats with systemic disease. However, many factors can affect the biostimulatory effects of LLLT. Thus, we attempted to investigate the effects of irradiation time on the healing of extraction sockets by evaluating the expressions of genes and proteins related to bone healing. The left and right first maxillary molars of 24 rats were extracted. Rats were randomly divided into four groups in which extraction sockets were irradiated for 0, 1, 2, or 5 min each day for 3 or 7 days. Specimens containing the sockets were examined using quantitative real-time reverse transcription polymerase chain reaction and western blotting. LLLT increased the expressions of all tested genes, Runx2, collagen type 1, osteocalcin, platelet-derived growth factor-B, and vascular endothelial growth factor, in a time-dependent manner. The highest levels of gene expressions were in the 5-min group after 7 days. Five minutes of irradiation caused prominent increases of the expression of all tested proteins after both 3 and 7 days. The expression level of each protein in group 4 was higher by almost twofold compared with group 1 after 7 days. Laser irradiation for 5 min caused the highest expressions of genes and proteins related to bone healing. In conclusion, LLLT had positive effects on the early stages of bone healing of extraction sockets in rats, which were irradiation time-dependent.


Assuntos
Lasers Semicondutores , Terapia com Luz de Baixa Intensidade , Alvéolo Dental/patologia , Alvéolo Dental/efeitos da radiação , Cicatrização/efeitos da radiação , Animais , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica , Masculino , Osteocalcina/genética , Osteocalcina/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo , Extração Dentária , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/genética
10.
Cell Physiol Biochem ; 34(5): 1824-34, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25502639

RESUMO

BACKGROUND/AIMS: Mussel-inspired polydopamine (PDA) is known to be an effective bioadhesive and bioactive material for controlling stem cell fate, which is important in stem cell-based regenerative medicine; however, the effect of PDA on osteogenic differentiation of periodontal ligament stem cells (PDLSCs) is not fully understood. In this study, we investigated the osteoinductive effect of PDA on PDLSCs and examined how this phenomenon is encouraged. METHODS: Osteogenic induction of PDLSCs was established by culturing cells on PDA film or on an uncoated polystyrene surface as a control. Osteogenic differentiation of PDLSCs was assessed by measurement of intracellular calcium levels and alkaline phosphatase (ALP) activity as well as by evaluation of protein expression of osteocalcin (OCN), osterix (OSX), and runt-related transcription factor 2 (RUNX2). RESULTS: The PDLSCs cultured on PDA film showed higher osteogenic activity than those on the control surface. Moreover, PDLSCs on PDA film expressed increased levels of the integrin adhesion receptors integrin α5 and ß1 compared to control cells. Expression of one isoform of the intracellular signaling protein phosphatidylinositol-3-kinase (PI3K), p110γ, was increased in PDLSCs on PDA film in a PDA dose-dependent manner. This signaling protein was found to interact with integrin ß1, demonstrating integrin-linked PI3K activation in response to PDA. Finally, the blockage of PI3K reduced the PDA-induced osteogenic activity of PDLSCs. CONCLUSION: our findings suggest that the bioadhesive PDA stimulates osteogenic differentiation of PDLSCs via activation of the integrin α5/ß1 and PI3K signaling pathways.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Indóis/farmacologia , Integrinas/metabolismo , Osteogênese/efeitos dos fármacos , Ligamento Periodontal/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Polímeros/farmacologia , Células-Tronco/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Osteocalcina/metabolismo , Osteocalcina/fisiologia , Osteogênese/fisiologia , Ligamento Periodontal/metabolismo , Ligamento Periodontal/fisiologia , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/fisiologia
11.
Cell Biochem Funct ; 32(7): 605-11, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25187163

RESUMO

Human periodontal ligament-derived stem cells (PDLSCs) demonstrate self-renewal capacity and multilineage differentiation potential. In this study, we investigated the transdifferentiation potential of human PDLSCs into pancreatic islet cells. To form three-dimensional (3D) clusters, PDLSCs were cultured in Matrigel with media containing differentiation-inducing agents. We found that after 6 days in culture, PDLSCs underwent morphological changes resembling pancreatic islet-like cell clusters (ICCs). The morphological characteristics of PDLSC-derived ICCs were further assessed using scanning electron microscopy analysis. Using reverse transcription-polymerase chain reaction analysis, we found that pluripotency genes were downregulated, whereas early endoderm and pancreatic differentiation genes were upregulated, in PDLSC-derived ICCs compared with undifferentiated PDLSCs. Furthermore, we found that PDLSC-derived ICCs were capable of secreting insulin in response to high concentrations of glucose, validating their functional differentiation into islet cells. Finally, we also performed dithizone staining, as well as immunofluorescence assays and fluorescence-activated cell sorting analysis for pancreatic differentiation markers, to confirm the differentiation status of PDLSC-derived ICCs. These results demonstrate that PDLSCs can transdifferentiate into functional pancreatic islet-like cells and provide a novel, alternative cell population for pancreatic repair.


Assuntos
Transdiferenciação Celular , Ilhotas Pancreáticas/citologia , Ligamento Periodontal/citologia , Células-Tronco/citologia , Linhagem da Célula , Células Cultivadas , Endoderma/citologia , Endoderma/metabolismo , Humanos , Ilhotas Pancreáticas/metabolismo , Dente Molar/citologia , Células-Tronco/metabolismo
12.
Toxicol Appl Pharmacol ; 259(3): 329-37, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22285274

RESUMO

Sodium fluoride (NaF) is used as a source of fluoride ions in diverse applications. Fluoride salt is an effective prophylactic for dental caries and is an essential element required for bone health. However, fluoride is known to cause cytotoxicity in a concentration-dependent manner. Further, no information is available on the effects of NaF on mouse embryonic stem cells (mESCs). We investigated the mode of cell death induced by NaF and the mechanisms involved. NaF treatment greater than 1mM reduced viability and DNA synthesis in mESCs and induced cell cycle arrest in the G(2)/M phase. The addition of NaF induced cell death mainly by apoptosis rather than necrosis. Catalase (CAT) treatment significantly inhibited the NaF-mediated cell death and also suppressed the NaF-mediated increase in phospho-c-Jun N-terminal kinase (p-JNK) levels. Pre-treatment with SP600125 or z-VAD-fmk significantly attenuated the NaF-mediated reduction in cell viability. In contrast, intracellular free calcium chelator, but not of sodium or calcium ion channel blockers, facilitated NaF-induced toxicity in the cells. A JNK specific inhibitor (SP600125) prevented the NaF-induced increase in growth arrest and the DNA damage-inducible protein 45α. Further, NaF-mediated loss of mitochondrial membrane potential was apparently inhibited by pifithrin-α or CAT inhibitor. These findings suggest that NaF affects viability of mESCs in a concentration-dependent manner, where more than 1mM NaF causes apoptosis through hydroxyl radical-dependent and caspase- and JNK-mediated pathways.


Assuntos
Apoptose/efeitos dos fármacos , Cariostáticos/toxicidade , Células-Tronco Embrionárias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Fluoreto de Sódio/toxicidade , Animais , Cariostáticos/administração & dosagem , Catalase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , DNA/biossíntese , DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Radical Hidroxila/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Fluoreto de Sódio/administração & dosagem
13.
J Cell Biochem ; 112(7): 1880-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21433060

RESUMO

Wnt/ß-catenin signaling regulates various cellular events involved in the proliferation and differentiation and these events are affected sensitively by applying to mechanical stimuli. However, the mechanisms by which mechanical force stimulates cardiomyogenesis are not extensively explored. In this study we investigated the cellular mechanisms by which ß-catenin signaling regulates cardiac differentiation of strain-subjected embryonic stem (ES) cells. The application of cells to cyclic strain increased beating cardiomyocyte foci with the attendant increases of Cx 43 and Nkx 2.5 proteins. Anti-oxidants such as vitamin C or N-acetyl cysteine (NAC) blocked the strain-mediated increases of Cx 43, Nkx 2.5, and α5/ß1 integrins. These anti-oxidants also suppressed the activation of phosphoinositide 3-kinase (PI3K) and Akt in cyclic strain-subjected cells. Western blot analysis revealed that PI3K is a critical downstream effector of ß1 integrin signaling and mediates Cx 43 and Nkx 2.5 expression in cyclic strain-applied ES cells. Cyclic strain increased the expression of ß-catenin and stimulated its nuclear translocation from the cytosol, which was prevented by anti-oxidant treatment. In addition, the application to cyclic strain increased mRNA expression of ß-catenin target genes, Axin2 and c-myc, as well as the phosphorylation of glycogen synthase kinase-3ß. Furthermore, the blockage of ß-catenin by its specific siRNA transfection diminished the cellular levels of Cx 43 and Nkx 2.5 proteins and the number of beating cardiomyocyte foci. Collectively, these results suggest that ß-catenin-mediated signaling is required for cyclic strain-stimulated cardiomyogenesis through ROS-dependent and integrin-mediated PI3K-Akt signaling cascades.


Assuntos
Células-Tronco Embrionárias/citologia , Integrina alfa5beta1/metabolismo , Miócitos Cardíacos/citologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , beta Catenina/metabolismo , Acetilcisteína/farmacologia , Androstadienos/farmacologia , Animais , Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Conexina 43/metabolismo , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Periodicidade , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais , Estresse Mecânico , Fatores de Transcrição/metabolismo , Wortmanina , beta Catenina/genética
14.
Biomacromolecules ; 12(4): 1224-33, 2011 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-21344942

RESUMO

A biocompatible, robust polymer micelle bearing pH-hydrolyzable shell cross-links was developed for efficient intracellular delivery of doxorubicin (DOX). The rationally designed triblock copolymer of poly(ethylene glycol)-poly(L-aspartic acid)-poly(L-phenylalanine) (PEG-PAsp-PPhe) self-assembled to form polymer micelles with three distinct domains of the PEG outer corona, the PAsp middle shell, and the PPhe inner core. Shell cross-linking was performed by the reaction of ketal-containing cross-linkers with Asp moieties in the middle shells. The shell cross-linking did not change the micelle size and the spherical morphology. Fluorescence quenching experiments confirmed the formation of shell cross-linked diffusion barrier, as judged by the reduced Stern-Volmer quenching constant (K(SV)). Dynamic light scattering and fluorescence spectroscopy experiments showed that shell cross-linking improved the micellar physical stability even in the presence of micelle disrupting surfactants, sodium dodecyl sulfate (SDS). The hydrolysis kinetics study showed that the hydrolysis half-life (t(1/2)) of ketal cross-links was estimated to be 52 h at pH 7.4, whereas 0.7 h at pH 5.0, indicating the 74-fold faster hydrolysis at endosomal pH. Ketal cross-linked micelles showed the rapid DOX release at endosomal pH, compared to physiological pH. Confocal laser scanning microscopy (CLSM) showed that ketal cross-linked micelles were taken up by the MCF-7 breast cancer cells via endocytosis and transferred into endosomes to hydrolyze the cross-links by lowered pH and finally facilitate the DOX release to inhibit proliferation of cancer cells. This ketal cross-linked polymer micelle is promising for enhanced intracellular delivery efficiency of many hydrophobic anticancer drugs.


Assuntos
Aminoácidos/química , Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Micelas , Polietilenoglicóis/química , Polímeros/química , Linhagem Celular Tumoral , Cromatografia em Gel , Portadores de Fármacos , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Cinética , Espectroscopia de Ressonância Magnética , Dodecilsulfato de Sódio/química , Espectrometria de Fluorescência
15.
J Nanosci Nanotechnol ; 11(7): 5689-97, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22121592

RESUMO

In this study, poly(D,L lactic-co-glycolic acid) (PLGA) was used as a drug carrier to generate two types of stents loaded with different concentrations of sirolimus. These stents were prepared by ultrasonic atomizing spray coating. Ultrasonic atomizing spray nozzle uses a low-pressure air/gas to produce a soft, highly focused beam of small spray drops. An isolated hypotube delivers liquid to the nozzle's atomizing surface while air/gas, delivered through the nozzle orifice at a fixed low pressure, shapes the atomized drops into a very precise, targeted spray. The stent was moved both in the traverse direction and rotated during the spraying process. The morphology of the sirolimus-eluting stents was examined by scanning electron microscopy (SEM) which indicated that the coating was very smooth and uniform. The coating was found to have the ability to withstand the compressive and tensile strains imparted without cracking during the stent inflation process. Release profile of sirolimus was measured by high performance liquid chromatography (HPLC). The release behavior of sirolimus from the stent surface had a two phase release profile with a burst release period of about 2 days, followed by a sustained and slow release phase. The mass loss behavior of PLGA appeared linear throughout most of the degradation period. At 28 days, neointimal formation was found to be significantly decreased for both sirolimus-eluting stents as compared to bare-metal stents (BMS). Assessment of vascular healing revealed an absence of increased inflammation in both sirolimus-eluting stents. Inflammation is commonly observed in drug-eluting stents (DES) with nonbiodegradable polymeric coatings. Taking these results into account, these novel sirolimus-eluting stents may be good candidates to resolve in-stent restenosis.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Stents Farmacológicos , Sirolimo/administração & dosagem , Análise de Variância , Animais , Cromatografia Líquida de Alta Pressão , Materiais Revestidos Biocompatíveis/química , Ácido Láctico/química , Teste de Materiais , Microscopia Eletrônica de Varredura , Nebulizadores e Vaporizadores , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Desenho de Prótese , Sirolimo/química , Sirolimo/farmacocinética , Suínos , Água
16.
J Nanosci Nanotechnol ; 11(7): 6371-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22121718

RESUMO

Electrospun Nanofiber sheets have been shown to mimic the structure of extracellular matrix (ECM). Although these nanofibers have shown great potential for use as tissue engineering scaffolds, it is difficult for the electrospun nanofiber based sheets to be shaped into the desired three-dimensional structure. In this study, poly(L-lactic acid) (PLLA), a biodegradable and biocompatible polyester, was electrospun to produce nanofibers that were treated with an amino group containing base in order to fabricate polymeric nanocylinders. The aspect ratio of the PLLA nanocylinders was tunable by varying the aminolysis time and density of the amino group containing base. The effects of changes in nanofibrous morphology of the PLLA nanocylinders/macro-porous gelatin scaffolds on cell adhesion and proliferation were evaluated. The results revealed different cell morphology, adhesion, and proliferation in the nanocylinders composite gelatin scaffold versus gelatin scaffold alone. Confocal laser scanning microscopy observation showed more spreading and a more flattened cell morphology after NIH3T3 cells were cultured on PLLA nanocylinders/gelatin scaffolds for 10 hours and 4 days. These results indicate that the gelatin/PLLA nanocylinder composite is a promising way to fabricate 3D nanofibrous scaffolds that accelerates cell adhesion and proliferation for tissue engineering.


Assuntos
Gelatina/química , Ácido Láctico/química , Nanotubos/química , Polímeros/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ácido Láctico/farmacologia , Teste de Materiais , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Células NIH 3T3 , Nanofibras , Nanotecnologia , Poliésteres , Polímeros/farmacologia , Porosidade , Espectroscopia de Infravermelho com Transformada de Fourier
17.
Artigo em Inglês | MEDLINE | ID: mdl-33919535

RESUMO

Dental disease is one of the most prevalent chronic diseases worldwide, and its expenditure is continuously increasing. Periodontal disease is increasing as a chronic non-communicable disease in adults and older people. Health screening has been shown to be cost-effective and improves the quality of life through the early detection of diseases. This study aimed to analyze the relationship between national health screening and dental scaling as a preventive service for periodontal disease. The study used sample cohort data from 2002 to 2015 provided by the National Health Insurance Sharing Service in South Korea. A logistic regression analysis of the utilization of dental scaling was performed to identify the independent effects of national health screening. People who underwent health screening showed a higher tendency to undergo dental scaling. Additionally, disparities in utilization according to socioeconomic status were reduced among those who underwent screening. The intervention to extend dental coverage could be more beneficial when combined with health screening, encouraging more people to participate and reducing inequalities in utilization.


Assuntos
Doenças Periodontais , Qualidade de Vida , Adulto , Idoso , Raspagem Dentária , Humanos , Serviços Preventivos de Saúde , República da Coreia/epidemiologia
18.
Tissue Cell ; 66: 101390, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32933713

RESUMO

This study investigated the delivery of S-nitrosothiol (GSNO) as a nitric oxide (NO) donor loaded into calcium carbonate-based mineralized nanoparticles (GSNO-MNPs) to regulate cell signaling pathways for the osteogenic differentiation of mouse embryonic stem cells (ESCs). GSNO-MNPs were prepared by an anionic block copolymer template-mediated calcium carbonate (CaCO3) mineralization process in the presence of GSNO. GSNO-MNPs were spherical and had a narrow size distribution. GSNO was stably loaded within the MNPs without denaturation. TEM analysis also demonstrated the localization of GSNO-MNPs within membrane-bound structures in the cell, indicating the successful introduction of GSNO-MNPs into the cytosol of ESCs. Intracellular levels of NO and cGMP were significantly increased upon treatment with GSNO-MNPs, compared with the control group. When cells were exposed to GSNO-MNPs, the effects of nanoparticles on cell viability were not statistically significant. GSNO-MNPs treatment increased ALP activity assay and intracellular calcium levels. Real-time RT-PCR also revealed highly increased expression levels of the osteogenic target genes ALP, osteocalcin (OCN), and osterix (OSX) in GSNO-MNP-treated ESCs. The protein levels of OSX and Runt-related transcription factor 2 (RUNX2) showed similar patterns of expression based on real-time RT-PCR. These results indicate that GSNO-MNPs influenced the osteogenic differentiation of ESCs. Transcriptome profiling identified several significantly enriched and involved biological networks, such as RAP1, RAS, PI3K-AKT, and MAPK signaling pathways. These findings suggest that GSNO-MNPs can modulate osteogenic differentiation in ESCs via complex molecular pathways.


Assuntos
Carbonato de Cálcio/química , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/citologia , Nanopartículas/química , Óxido Nítrico/farmacologia , Osteogênese/efeitos dos fármacos , Animais , Diferenciação Celular/genética , Perfilação da Expressão Gênica , Camundongos , Minerais/química , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Nanopartículas/ultraestrutura , Osteogênese/genética , RNA-Seq , S-Nitrosotióis/farmacologia
19.
J Cell Biochem ; 106(6): 1060-7, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19206162

RESUMO

Type I collagen (COL I) is the predominant collagen in the extracellular matrix of periodontal ligament (PDL), and its expression in PDL fibroblasts (PLF) is sensitive to mechanical force. However, the mechanism by which PLF induces COL I to respond to mechanical force is unclear. This study examined the nature of human PLF in mediating COL I expression in response to centrifugal force. Signal transduction pathways in the early stages of mechanotransduction involved in the force-driven regulation of COL I expression were also investigated. Centrifugal force up-regulated COL I without cytotoxicity and activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 kinase. ERK and JNK inhibitor blocked the expression of COL I but p38 kinase inhibitor had no effect. Centrifugal force activated activator protein-1 (AP-1) through dimerization between c-Fos and c-Jun transcription factors. ERK and JNK inhibitors also inhibited AP-1-DNA binding, c-Fos nuclear translocation, and c-Jun phosphorylation that were increased in the force-exposed PLF. Further, transfecting the cells with c-Jun antisense oligonucleotides almost completely abolished the force-induced increase of c-Jun phosphorylation and COL I induction. Our findings suggest that mechanical signals are transmitted into the nucleus by ERK/JNK signaling pathways and then stimulate COL I expression through AP-1 activation in force-exposed human PLF.


Assuntos
Colágeno Tipo I/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ligamento Periodontal/citologia , Estresse Mecânico , Fator de Transcrição AP-1/metabolismo , Adulto , Células Cultivadas , Colágeno Tipo I/genética , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , Fibroblastos/citologia , Regulação da Expressão Gênica , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Fator de Transcrição AP-1/genética , Adulto Jovem , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
J Cell Biochem ; 106(5): 787-97, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19199341

RESUMO

Reactive oxygen species (ROS) generated by a variety of endogenous factors and roles in embryonic stem (ES) cells has yet to be identified. Thus, we examined role of arachidonic acid (AA) in H(2)O(2)-induced proliferation of mouse ES cells and its related signaling molecules. AA release was maximally increased in response to 10(-4) M H(2)O(2) for 1 h. In addition, H(2)O(2) increased intracellular Ca(2+) concentration ([Ca(2+)](i)) and the phosphorylation of protein kinase C (PKC), p44/42, p38 mitogen-activated protein kinase (MAPK), and JNK/SAPK. Moreover, H(2)O(2) induced an increase in the phosphorylation of epidermal growth factor receptor (EGFR), which was blocked by the inhibition of p44/42 or p38 MAPKs. The inhibition of each signal molecule with specific inhibitors blocked H(2)O(2)-induced cytosolic phospholipase A(2) (cPLA(2)) activation and AA release. H(2)O(2) increased NF-kappaB phosphorylation to induce an increase in the levels of cyclooxygenase (COX)-2 proteins. Subsequently, H(2)O(2) stimulated PGE(2) synthesis, which was reduced by the inhibition of NF-kappaB activation. Moreover, each H(2)O(2) or PGE(2) increased DNA synthesis and the number of cells. However, H(2)O(2)-induced increase in DNA synthesis was inhibited by the suppression of cPLA(2) pathway. In conclusion, H(2)O(2) increased AA release and PGE(2) production by the upregulation of cPLA(2) and COX-2 via Ca(2+)/PKC/MAPKs and EGFR transactivation, subsequently proliferation of mouse ES cells.


Assuntos
Ácido Araquidônico/metabolismo , Proliferação de Células , Ciclo-Oxigenase 2/genética , Células-Tronco Embrionárias/citologia , Receptores ErbB/genética , Fosfolipases A2/genética , Ativação Transcricional , Animais , Cálcio/metabolismo , Dinoprostona/biossíntese , Peróxido de Hidrogênio/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA