Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Dent Mater J ; 43(2): 191-199, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38246630

RESUMEN

Gingival epithelial attachment to the abutment is important for the prevention of peri-implantitis. Polyetheretherketone (PEEK) has recently gained attention as an alternative material to titanium; however, it is biologically inert, which is disadvantageous for obtaining soft tissue sealing of the transmucosal part of the implant abutment. Therefore, ultraviolet (UV) irradiation, argon plasma irradiation, and buffing were selected as treatments to modify the PEEK surface. None of the treatments had any effect on the material's mechanical strength. The UV and plasma treatments did not significantly affect the surface morphology. Surface elemental analysis showed a decrease in carbon content and an increase in oxygen content and wettability for all treatments. Human gingival epithelial cell adhesion, proliferation, and the expression of adhesion proteins integrin ß4 and laminin 332, were increased. Surface modification to PEEK was suggested to enhance cell activity on PEEK.


Asunto(s)
Benzofenonas , Polietilenglicoles , Polímeros , Humanos , Propiedades de Superficie , Cetonas , Adhesión Celular , Titanio , Células Epiteliales
3.
Am J Med ; 137(3): 273-279.e2, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37984772

RESUMEN

BACKGROUND: We determined the effects and an accurate marker of periodontal treatment on serum interleukin (IL)-6 and high-sensitivity C-reactive protein (HsCRP) levels in systemically healthy individuals with periodontal disease. METHODS: This multicenter study included systemically healthy individuals with periodontal disease who received initial periodontal treatment and had no periodontal treatment history. Periodontal parameters, including periodontal inflamed surface area, masticatory efficiency, and periodontal disease classification; serum IL-6 and HsCRP levels; and serum immunoglobulin (Ig)G titers against periodontal pathogens were evaluated at baseline and after treatment. Subjects were classified as low or high responders (group) based on periodontal inflamed surface area changes. RESULTS: There were 153 participants. Only periodontal inflamed surface area changes were markedly different between low and high responders. Periodontal treatment (time point) decreased both serum IL-6 and HsCRP levels. The interaction between group and time point was remarkable only for serum IL-6 levels. Changes in serum immunoglobulin (Ig)G titers against periodontal pathogens were not associated with IL-6 changes in high responders. We analyzed the indirect effect of serum anti-Porphyromonas gingivalis type 2 IgG titer changes using mediation analysis and found no significance. However, the direct effect of group (low or high responder) on IL-6 changes was considerable. CONCLUSIONS: Periodontal treatment effectively decreased serum IL-6 levels, independent of periodontal pathogen infection, in systemically healthy individuals with periodontal disease.


Asunto(s)
Proteína C-Reactiva , Enfermedades Periodontales , Humanos , Proteína C-Reactiva/análisis , Interleucina-6 , Inflamación , Enfermedades Periodontales/terapia , Inmunoglobulinas
4.
Sci Rep ; 13(1): 17081, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37816801

RESUMEN

Photodynamic therapy is a treatment that combines a light source with a photosensitizer. LEDs have attracted considerable attention in clinical dentistry because they are inexpensive and safe to use. Although the interaction between photosensitizers and LEDs in dental practice is effective for treating periodontal disease by killing periodontopathic bacteria, little is known about the effects of LEDs on human gingival fibroblasts (HGnFs), which play an important role in gingival wound healing. In this study, we investigated the effects of high-intensity red LED irradiation on HGnFs after the addition of methylene blue (MB), one of the least harmful photosensitizers, on wound healing and reactive oxygen species (ROS) production induced by photodynamic reactions. We found that irradiation of MB with high-intensity red LED at controlled energy levels promoted cell proliferation, migration, and production of wound healing factors. Furthermore, ROS production by a photodynamic reaction enabled the translocation of phosphorylated Grb2-associated binder-1, activating Extracellular signal-regulated kinase 1/2 and c-Jun N-terminal kinase signals. Our findings suggest that proper control of ROS production has a beneficial effect on gingival fibroblasts, which constitute periodontal tissue, from the perspective of gingival wound healing.


Asunto(s)
Fotoquimioterapia , Fármacos Fotosensibilizantes , Humanos , Especies Reactivas de Oxígeno/farmacología , Fármacos Fotosensibilizantes/farmacología , Encía , Cicatrización de Heridas , Azul de Metileno/farmacología
5.
Int J Mol Sci ; 24(20)2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37894999

RESUMEN

Intracellular nutrient metabolism, particularly the metabolism of essential amino acids (EAAs), is crucial for cellular functions, including energy production and redox homeostasis. An EAA deficiency can lead to cellular dysfunction and oxidative stress. This study explores the mechanisms underlying cellular responses to EAA starvation, focusing on ROS-induced DNA damage and apoptosis. MC3T3-E1 cells were subjected to EAA starvation, and various assays were conducted to assess cell proliferation, survival, DNA damage, and apoptosis. The antioxidant N-acetylcysteine (NAC) was employed to block ROS formation and mitigate cellular damage. Gene expression and Western blot analyses were performed to elucidate molecular pathways. EAA starvation-induced ROS generation, DNA damage, and apoptosis in MC3T3-E1 cells. NAC administration effectively reduced DNA damage and apoptosis, highlighting the pivotal role of ROS in mediating these cellular responses during EAA deficiency. This study demonstrates that EAA starvation triggers ROS-mediated DNA damage and apoptosis, offering insights into the intricate interplay between nutrient deficiency, oxidative stress, and programmed cell death. NAC emerges as a potential therapeutic intervention to counteract these adverse effects.


Asunto(s)
Apoptosis , Estrés Oxidativo , Ratones , Animales , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Acetilcisteína/metabolismo , Daño del ADN , Osteoblastos/metabolismo , Aminoácidos Esenciales/metabolismo
6.
Biochem Biophys Res Commun ; 672: 168-176, 2023 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-37354610

RESUMEN

This study investigates the effects of essential amino acid (EAA) starvation on murine osteoblasts cells and the underlying mechanisms. We performed and observed the cell proliferation, autophagy, and osteogenic differentiation under deprivation of EAA in vitro. The results showed that EAA starvation resulted in cell cycle arrest via phosphorylation of the MAPK signaling pathway, leading to inhibition of cell proliferation and osteogenic differentiation. Additionally, the LKB1-AMPK signaling pathway was also found to be phosphorylated, inducing autophagy. These findings highlight the significant role of EAA in regulating cellular processes. Furthermore, this study contributes to our understanding of the effects of nutrient deprivation on cellular physiology and may aid in the development of novel therapeutic strategies for diseases associated with amino acid metabolism.


Asunto(s)
Autofagia , Osteogénesis , Animales , Ratones , Diferenciación Celular , Aminoácidos Esenciales/metabolismo , Aminoácidos Esenciales/farmacología , Puntos de Control del Ciclo Celular , Osteoblastos/metabolismo
7.
J Periodontal Res ; 58(3): 607-620, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36883427

RESUMEN

BACKGROUND: Periodontal ligament cells (PDLCs), as mesenchymal cells in the oral cavity, are closely linked to periodontal tissue regeneration. However, the effect of local glucose deficiency on periodontal tissue regeneration, such as immediately post-surgery, remains unknown. OBJECTIVE: In the present study, we investigated the effect of a low-glucose environment on the proliferation and osteogenic differentiation of PDLCs. MATERIALS AND METHODS: We used media with five glucose concentrations (100, 75, 50, 25, and 0 mg/dL) and focused on the effects of a low-glucose environment on the proliferation, osteogenic differentiation, and autophagy of PDLCs. Additionally, we focused on changes in lactate production in a low-glucose environment and investigated the involvement of lactate with AZD3965, a monocarboxylate transporter-1 (MCT-1) inhibitor. RESULTS: The low-glucose environment inhibited PDLCs proliferation, migration, and osteogenic differentiation, and induced the expression of the autophagy-related factors LC3 and p62. Lactate and ATP production were decreased under low-glucose conditions. The addition of AZD3965 (MCT-1 inhibitor) in normal glucose conditions caused a similar trend as in low-glucose conditions on PDLCs. CONCLUSION: Our results suggest lactate production through glucose metabolism in the osteogenic differentiation of PDLCs. A low-glucose environment decreased lactate production, inhibiting cell proliferation, migration, and osteogenic differentiation and inducing autophagy in PDLCs.


Asunto(s)
Osteogénesis , Ligamento Periodontal , Humanos , Células Cultivadas , Diferenciación Celular , Proteínas Portadoras/metabolismo , Proliferación Celular , Lactatos/metabolismo , Lactatos/farmacología
8.
Antioxidants (Basel) ; 11(10)2022 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-36290630

RESUMEN

Gingival tissue experiences an environment of nutrient shortage, such as low glucose conditions, after periodontal surgery. Our previous studies found that this low glucose condition inhibits normal gingival cell functions. However, the mechanism by which this glucose-deficient environment causes cellular damage to human gingival fibroblasts (HGnFs) remains unclear. This study aimed to investigate the biological effects of ROS induction on HGnFs under low glucose conditions. ROS levels and cellular anti-ROS ability of HGnFs under different glucose concentrations were evaluated by measuring ROS formation and the expression of superoxide dismutase and heme oxygenase 1. Changes in cellular viability were investigated using 5-bromo-2'-deoxyuridine assay and cell survival detection, and the cellular damage was evaluated by the expression of inflammatory cytokines and changes in the expression of autophagy-related protein. ROS formation was then blocked using N-acetyl-L-cysteine (NAC), and the effects of ROS on HGnFs under low glucose conditions were investigated. Low glucose conditions induced ROS accumulation, reduced cellular activity, and induced inflammation and autophagy. After NAC application, the anti-ROS capacity increased, cellular activity improved, and inflammation and autophagy were controlled. This can be effectively controlled by the application of antioxidants such as NAC.

9.
Life (Basel) ; 12(5)2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35629403

RESUMEN

Periodontitis is an inflammatory lesion in the periodontal tissue. The behavior of human periodontal ligament stem cells (hPDLSCs), which play an important role in periodontal tissue regeneration, is restricted by the influence of inflammatory mediators. Photobiomodulation therapy exerts anti-inflammatory effects. The purpose of this study was to investigate the effects of light-emitting diode (LED) irradiation on the inflammatory responses of hPDLSCs. The light source was a red LED (peak wavelength: 650 nm), and the total absolute irradiance was 400 mW/cm2. The inflammatory response in hPDLSCs is induced by tumor necrosis factor (TNF)-α. Adenosine triphosphate (ATP) levels and pro-inflammatory cytokine (interleukin [IL]-6 and IL-8) production were measured 24 h after LED irradiation, and the effects of potassium cyanide (KCN) were investigated. LED irradiation at 6 J/cm2 significantly increased the ATP levels and reduced TNF-α-induced IL-6 and IL-8 production. Furthermore, the inhibitory effect of LED irradiation on the production of pro-inflammatory cytokines was inhibited by KCN treatment. The results of this study showed that high-intensity red LED irradiation suppressed the TNF-α-stimulated pro-inflammatory cytokine production in hPDLSCs by promoting ATP synthesis. These results suggest that high-intensity red LED is a useful tool for periodontal tissue regeneration in chronically inflamed tissues.

10.
Sci Rep ; 12(1): 1230, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35075260

RESUMEN

Human gingival fibroblasts (HGnFs) maintain periodontal tissue homeostasis through active proliferation and migration. Clinically, it is considered that the wound-healing ability of the gingival tissue is maintained even in environments with insufficient supply of nutrients, such as glucose, immediately after periodontal surgery. However, the effects of such glucose-deficient environments on HGnFs remain unclear. This study aimed to investigate the effects of low-glucose environment on HGnFs homeostasis. We evaluated gingival wound healing by examining cell proliferation and migration and collagen synthesis in HGnFs cultured in 100, 50, 25, and 0 mg/dL glucose in vitro. The cellular stress levels were determined by measuring the lactate dehydrogenase (LDH) and reactive oxygen species (ROS) levels. The glucose metabolism of HGnFs in the low-glucose concentrations was studied by measuring glucose transporter type 1 (GLUT1) mRNA expression, glucose uptake assays, lactate and ATP productions. Molecular effects were examined with a focus on the LKB1-AMPK signaling pathway. Autophagy activity in glucose-deprived HGnFs was evaluated by measuring the levels of autophagy-related proteins. Low glucose levels increased cellular stress levels, autophagy activity, and enhanced glucose metabolism through the LKB1-AMPK signaling pathway, providing more ATPs to promote wound healing. Our results regarding glucose transfer suggest the rapid healing of gingival wounds.


Asunto(s)
Autofagia , Fibroblastos/fisiología , Encía/fisiología , Glucosa/deficiencia , Cicatrización de Heridas , Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Células Cultivadas , Glucólisis , Humanos
11.
Lasers Med Sci ; 36(1): 55-65, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32588268

RESUMEN

Photobiomodulation therapy (PBMT) using a light-emitting diode (LED) has been employed for various photomedicine studies. The aim of this study was to determine the effects of a high-intensity red LED on the proliferation and osteogenic differentiation of human bone marrow mesenchymal stem cells (BMSCs) and the related mechanism. BMSCs were subjected to high-intensity red LED (LZ1-00R205 Deep Red LED) irradiations for 0 to 40 s with energy densities ranging from 0 to 8 J/cm2. The distance from the LED to the cell layer was 40 mm. The spot size on the target was 4 cm2. Cell proliferation was measured at 3, 24, 48, and 72 h. The effects of LED irradiation on osteogenic differentiation and mineralization were examined with a particular focus on the Wnt/ß-catenin signaling pathway. The high-intensity red LED irradiations did not alter BMSC proliferation after 72 h. LED exposure of 6 J/cm2 (30 s) led to significant enhancements of osteogenic differentiation and mineralization. Additionally, the high-intensity LED irradiation induced activation of Wnt/ß-catenin. The effects of the high-intensity LED irradiation on BMSC osteogenic differentiation and mineralization were suppressed by treatment with the Wnt/ß-catenin inhibitor XAV939. P < 0.05 was considered significant. The results indicate that high-intensity red LED irradiation increases BMSC osteogenic differentiation and mineralization via Wnt/ß-catenin activation. Therefore, short duration irradiation with a portable high-intensity LED may be used as a potential approach in hard tissue regeneration therapy.


Asunto(s)
Calcificación Fisiológica/efectos de la radiación , Diferenciación Celular/efectos de la radiación , Luz , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de la radiación , Osteogénesis/efectos de la radiación , Vía de Señalización Wnt/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Cultivadas , Humanos
12.
Molecules ; 24(19)2019 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-31574951

RESUMEN

Shikonin, an active ingredient of Lithospermum erythrorhizon, exerts anti-inflammatory and antibacterial effects, and promotes wound healing. We investigated whether shikonin stimulated gingival tissue wound healing in human gingival fibroblasts (hGF). In addition, we evaluated the effects of shikonin on the mitogen-activated protein kinase (MAPK) signaling pathway, which has an important role in wound healing. hGF were subjected to primary culture using gingiva collected from patients. The cells were exposed to/treated with Shikonin at concentrations ranging from 0.01 to 100 µM. The optimal concentration was determined by cell proliferation and migration assays. Type I collagen and fibronectin synthesis, the gene expression of vascular endothelial growth factor (VEGF) and FN, and the phosphorylation of Extracellular signal-regulated kinase (ERK) 1/2 were investigated. Identical experiments were performed in the presence of PD98059 our data suggest, a specific ERK 1/2 inhibitor. Shikonin significantly promoted hGF proliferation and migration. Shikonin (1 µM) was chosen as the optimal concentration. Shikonin promoted type I collagen and FN synthesis, increased VEGF and FN expression, and induced ERK 1/2 phosphorylation. These changes were partially suppressed by PD98059. In conclusion, Shikonin promoted the proliferation, migration, type I collagen and FN synthesis, and expression of VEGF and FN via ERK 1/2 signaling pathway in hGFs. Therefore, shikonin may promote periodontal tissue wound healing.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Fibroblastos/metabolismo , Encía/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Naftoquinonas/farmacología , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Fibronectinas/metabolismo , Expresión Génica , Humanos , Lactato Deshidrogenasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/efectos de los fármacos
13.
BMC Oral Health ; 19(1): 188, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31426773

RESUMEN

BACKGROUND: We previously showed that nasal administration of a combination of dendritic cell (DC) targeted DNA plasmid expressing Flt3 ligand and CpG oligodeoxynucleotides 1826 as a mucosal adjuvant (double adjuvant, DA) provoked protective immunity in the upper respiratory tract of young adult and aging mice. Here, we investigated whether the nasal DA system induces secretory (S)IgA antibodies (Abs) toward recombinant fimbrillin (rFimA) of Porphyromonas gingivalis (P. gingivalis) in the saliva of young adult and aging mice. Further, we examined the functional applicability of rFimA-specific salivary SIgA Abs. METHODS: BALB/c mice (8- or 48-week-old) were nasally immunized with rFimA plus DA three times at weekly intervals. Control mice were nasally administered rFimA alone. Saliva samples were collected 1 week after the final immunization, and were subjected to rFimA-specific ELISA. To examine the functional applicability of rFimA-specific SIgA Abs, IgA-enriched saliva samples were subjected to an inhibition assay in order to assess the numbers of P. gingivalis cells bound to the salivary protein statherin. RESULTS: The 8- and 48-week-old mice administered nasal rFimA plus DA showed significantly increased levels of rFimA-specific SIgA Abs in saliva and elevated numbers of CD11c+ DCs in sublingual glands (SLGs), periglandular lymph nodes (PGLNs) and submandibular glands (SMGs) as well as nasopharyngeal-associated lymphoid tissues (NALT) compared to mice administered rFimA alone. Further, rFimA-specific SIgA Abs-containing saliva, in which IgG Abs of 8- and 48-week-old mice administered nasal rFimA plus DA were removed, significantly inhibited binding of P. gingivalis to the salivary protein. CONCLUSIONS: These findings show that this DA system could be an effective nasal vaccine strategy for the enhancement of P. gingivalis-specific protective immunity in the oral cavity of adolescents and older individuals.


Asunto(s)
ADN , Inmunoglobulina A Secretora , Porphyromonas gingivalis , Proteínas y Péptidos Salivales , Animales , Humanos , Inmunidad , Ratones , Ratones Endogámicos BALB C , Proteínas y Péptidos Salivales/metabolismo , Vacunas de ADN
14.
J Oral Sci ; 60(4): 579-587, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30587690

RESUMEN

It is widely accepted that vitamin E (VE) acts as an antioxidant and is involved in various metabolic systems including the regulation of gene expression and inhibition of cell proliferation. The most predominant isoform of VE in the living body is α-tocopherol. However, the influence of α-tocopherol on bone marrow mesenchymal cells (BMMCs) in a background of type II diabetes mellitus (DM) has not been investigated. The focus of the present study was to clarify the effect of α-tocopherol on BMMCs derived from rats with type II DM and the underlying mechanisms involved. BMMCs were isolated from rats with type II DM. The BMMCs were either untreated or exposed to α-tocopherol at concentrations of 1.0, 10, and 100 µM, and the resulting effects of α-tocopherol on cell proliferation, H2O2 activity, and antioxidant and inflammatory cytokine production were examined. At 100 µM, α-tocopherol had no effect on cell proliferation, but H2O2 activity was significantly increased. At 10 µM, α-tocopherol increased the gene expression of IL-1ß, and markedly promoted that of TNF-α. Expression of catalase in the presence of 100 µM α-tocopherol was lower than for the other concentrations. At a low concentration, α-tocopherol exerted good antioxidant and anti-inflammatory effects on BMMCs. The study suggests that maintaining α-tocopherol at a low concentration might promote the recovery of BMMCs from oxidative stress.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Madre Mesenquimatosas/efectos de los fármacos , alfa-Tocoferol/farmacología , Animales , Western Blotting , Catalasa/metabolismo , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Peróxido de Hidrógeno/metabolismo , Estrés Oxidativo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
J Periodontol ; 89(3): 351-360, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29528486

RESUMEN

BACKGROUND: Light-emitting diode (LED) is attracting attention as a new light source for phototherapy. However, its effects on periodontal tissue regeneration remain unknown. The aim of this study was to examine the effects of high-power, red LED irradiation on human periodontal ligament stem cells (PDLSCs), which play an important role in periodontal tissue regeneration. METHODS: PDLSCs were derived from adult human third molars. The light source was red LED (peak wavelength: 650 nm). Energy densities ranging from 0 to 10 J/cm2 were tested to determine the optimal dose. PDLSC proliferation was measured using two parameters: live cell protease and ATP levels. After the cells were induced to differentiate, the effect of LED irradiation on osteogenic differentiation and mineralization was examined, with particular focus on the extracellular signal-regulated kinase (ERK)1/2 signaling pathway using an ERK inhibitor (PD98059). RESULTS: LED irradiation at 8 J/cm2 led to a significant increase in PDLSC proliferation and enhanced Runx2 and Osterix mRNA expression, Alkaline phosphatase activity, procollagen type I C-peptide and osteocalcin production, calcium deposition, and alizarin red S staining. In addition, LED induced the activation of ERK1/2, and the effects of LED on PDLSC proliferation, differentiation, and mineralization could be suppressed by treatment with PD98059. CONCLUSIONS: The results of this study show that 650-nm high-power, red, LED irradiation increases PDLSCs proliferation, and osteogenic differentiation and mineralization, mediated by ERK1/2 activation. These findings suggest that LED may be a useful tool for periodontal tissue regeneration.


Asunto(s)
Osteogénesis , Ligamento Periodontal , Adulto , Fosfatasa Alcalina , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Transducción de Señal , Células Madre
16.
Int J Mol Sci ; 18(9)2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28925952

RESUMEN

We would like to submit the following correction to the published paper [1].[...].

17.
Int J Mol Sci ; 18(4)2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28383491

RESUMEN

In recent years, zirconia has been a recognized implant material in clinical dentistry. In the present study, we investigated the performance of an alkali-modified ceria-stabilized tetragonal ZrO2 polycrystalline ceramic-based nanostructured zirconia/alumina composite (NANOZR) implant by assessing surface morphology and composition, wettability, bovine serum albumin adsorption rate, rat bone marrow (RBM) cell attachment, and capacity for inducing bone differentiation. NANOZR surfaces without and with alkali treatment served as the control and test groups, respectively. RBM cells were seeded in a microplate with the implant and cultured in osteogenic differentiation medium, and their differentiation was evaluated by measuring alkaline phosphatase (ALP) activity, osteocalcin (OCN) production, calcium deposition, and osteogenic gene expression. The alkali-treated NANOZR surface increased ALP activity, OCN production, calcium deposition, and osteogenesis-related gene expression in attached RBM cells. These data suggest that alkali treatment enhances the osteogenesis-inducing capacity of NANOZR implants and may therefore improve their biointegration into alveolar bone.


Asunto(s)
Óxido de Aluminio/farmacología , Células de la Médula Ósea/citología , Materiales Dentales/farmacología , Osteogénesis/efectos de los fármacos , Circonio/farmacología , Óxido de Aluminio/química , Animales , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Implantes Dentales , Materiales Dentales/química , Nanopartículas/química , Ratas , Propiedades de Superficie , Humectabilidad , Circonio/química
18.
J Periodontol ; 87(4): e44-51, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26537370

RESUMEN

BACKGROUND: Diabetes mellitus (DM) is a major risk factor for periodontal disease and affects various cellular functions. Periodontal ligament stem cells (PDLSCs) play an important role in periodontal tissue regeneration; however, the effect of hyperglycemia on PDLSCs is unclear. The aim of this study is to investigate whether hyperglycemia affects periodontal tissue regeneration, using human PDLSCs and high-glucose medium as a model of DM. METHODS: PDLSCs were obtained from healthy adult human mandibular third molars. Cell proliferation, osteoblastic differentiation, and proinflammatory cytokine expression were investigated by culturing PDLSCs in media supplemented with four different glucose concentrations representative of control patients (5.5 mM), patients with postprandial or controlled DM (8.0 mM), and patients with uncontrolled DM (12.0 and 24.0 mM). The molecular effects of hyperglycemia on PDLSC physiology were examined with a focus on the nuclear factor (NF)-(κB signaling pathway. The involvement of NF-κB was investigated with a specific NF-κB inhibitor in PDLSCs under hyperglycemic conditions. RESULTS: High glucose levels inhibited PDLSC proliferation and differentiation into osteoblasts but induced NF-κB activation and subsequent interleukin (IL)-6 and IL-8 expression. Treatment with an NF-κB inhibitor rescued the defects in cell proliferation and osteoblastic differentiation and inhibited the IL-6 expression caused by the high-glucose environment. CONCLUSION: The results of this study demonstrate that hyperglycemia inhibits human PDLSC proliferation and osteoblastic differentiation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Osteoblastos , Ligamento Periodontal , Glucosa , Humanos , Células Madre
19.
J Periodontol ; 86(3): 448-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25494660

RESUMEN

BACKGROUND: Titanium (Ti) dental implants have been widely used for prosthetic reconstruction of dentition. Unfortunately, peri-implantitis can result in failure of dental implant osseointegration. Lipopolysaccharide (LPS) acts as a chronic inflammatory stimulus and maintains peri-implant inflammation, worsening the prognosis for implant osseointegration. The purpose of this study is to determine the effects of 10 M NaOH-modified Ti surface with nanonetwork structure on the proliferation and osteogenic differentiation of rat bone marrow mesenchymal stem cells (BMMSCs) in the context of Porphyromonas gingivalis LPS exposure. METHODS: Titanium disks treated with 10 M NaOH solution and control were incubated with BMMSCs and exposed to P. gingivalis LPS (0, 0.1, or 1 µg/mL). The effects of the modified nanonetwork structure on osteogenic differentiation of rat BMMSCs were evaluated in the context of different concentrations of P. gingivalis LPS exposure. RESULTS: Rat BMMSCs on the 10 M NaOH-modified Ti surface with nanonetwork structure had higher levels of osteogenesis-related gene expression and significantly greater cell proliferation, alkaline phosphatase activity, and extracellular matrix deposition and mineralization than cells on the untreated Ti surfaces, in all the groups with different doses of P. gingivalis LPS exposure. CONCLUSION: The 10 M NaOH-modified Ti surface with nanonetwork structure has better endotoxin tolerance under P. gingivalis LPS exposure than the non-modified surface.


Asunto(s)
Materiales Dentales/química , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Nanoestructuras/química , Osteogénesis/efectos de los fármacos , Porphyromonas gingivalis/fisiología , Titanio/química , Adsorción , Fosfatasa Alcalina/análisis , Animales , Calcificación Fisiológica/fisiología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/análisis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/análisis , Proteínas de la Matriz Extracelular/análisis , Sialoproteína de Unión a Integrina/análisis , Lipopolisacáridos/administración & dosificación , Células Madre Mesenquimatosas/fisiología , Osteocalcina/análisis , Osteonectina/análisis , Ratas , Ratas Sprague-Dawley , Hidróxido de Sodio/química , Propiedades de Superficie
20.
Int J Mol Sci ; 15(8): 14026-43, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25123134

RESUMEN

Enamel matrix derivative (EMD) is widely used in periodontal tissue regeneration therapy. However, because the bioactivity of EMD varies from batch to batch, and the use of a synthetic peptide could avoid use from an animal source, a completely synthetic peptide (SP) containing the active component of EMD would be useful. In this study an oligopeptide synthesized derived from EMD was evaluated for whether it contributes to periodontal tissue regeneration. We investigated the effects of the SP on cell proliferation and osteoblast differentiation of human mesenchymal stem cells (MSCs), which are involved in tissue regeneration. MSCs were treated with SP (0 to 1000 ng/mL), to determine the optimal concentration. We examined the effects of SP on cell proliferation and osteoblastic differentiation indicators such as alkaline phosphatase activity, the production of procollagen type 1 C-peptide and osteocalcin, and on mineralization. Additionally, we investigated the role of extracellular signal-related kinases (ERK) in cell proliferation and osteoblastic differentiation induced by SP. Our results suggest that SP promotes these processes in human MSCs, and that ERK inhibitors suppress these effects. In conclusion, SP promotes cell proliferation and osteoblastic differentiation of human MSCs, probably through the ERK pathway.


Asunto(s)
Esmalte Dental/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Oligopéptidos/síntesis química , Oligopéptidos/farmacología , Fosfatasa Alcalina/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Flavonoides/farmacología , Humanos , Oligopéptidos/química , Osteocalcina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...