Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
J Periodontal Res ; 59(3): 530-541, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38501357

RESUMEN

OBJECTIVE: The purpose of this study is to investigate regenerative process by immunohistochemical analysis and evaluate periodontal tissue regeneration following a topical application of BDNF to inflamed 3-wall intra-bony defects. BACKGROUND: Brain-derived neurotrophic factor (BDNF) plays a role in the survival and differentiation of central and peripheral neurons. BDNF can regulate the functions of non-neural cells, osteoblasts, periodontal ligament cells, endothelial cells, as well as neural cells. Our previous study showed that a topical application of BDNF enhances periodontal tissue regeneration in experimental periodontal defects of dog and that BDNF stimulates the expression of bone (cementum)-related proteins and proliferation of human periodontal ligament cells. METHODS: Six weeks after extraction of mandibular first and third premolars, 3-wall intra-bony defects were created in mandibular second and fourth premolars of beagle dogs. Impression material was placed in all of the artificial defects to induce inflammation. Two weeks after the first operation, BDNF (25 and 50 µg/mL) immersed into atelocollagen sponge was applied to the defects. As a control, only atelocollagen sponge immersed in saline was applied. Two and four weeks after the BDNF application, morphometric analysis was performed. Localizations of osteopontin (OPN) and proliferating cell nuclear antigen (PCNA)-positive cells were evaluated by immunohistochemical analysis. RESULTS: Two weeks after application of BDNF, periodontal tissue was partially regenerated. Immunohistochemical analyses revealed that cells on the denuded root surface were positive with OPN and PCNA. PCNA-positive cells were also detected in the soft connective tissue of regenerating periodontal tissue. Four weeks after application of BDNF, the periodontal defects were regenerated with cementum, periodontal ligament, and alveolar bone. Along the root surface, abundant OPN-positive cells were observed. Morphometric analyses revealed that percentage of new cementum length and percentage of new bone area of experimental groups were higher than control group and dose-dependently increased. CONCLUSION: These findings suggest that BDNF could induce cementum regeneration in early regenerative phase by stimulating proliferation of periodontal ligament cells and differentiation into periodontal tissue cells, resulting in enhancement of periodontal tissue regeneration in inflamed 3-wall intra-bony defects.


Asunto(s)
Pérdida de Hueso Alveolar , Factor Neurotrófico Derivado del Encéfalo , Cementogénesis , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Perros , Cementogénesis/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Osteopontina , Ligamento Periodontal/patología , Ligamento Periodontal/efectos de los fármacos , Masculino , Regeneración Tisular Guiada Periodontal/métodos , Regeneración Ósea/efectos de los fármacos , Cemento Dental/patología , Cemento Dental/efectos de los fármacos , Periodoncio/patología , Periodoncio/metabolismo , Mandíbula , Proliferación Celular/efectos de los fármacos
2.
Microsc Res Tech ; 87(7): 1584-1597, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38433562

RESUMEN

To evaluate the effects of premixed calcium silicate based ceramic sealers on the viability and osteogenic/cementogenic differentiation of human periodontal ligament stem cells (hPDLSCs). The materials evaluated were TotalFill BC Sealer (TFbc), AH Plus Bioceramic Sealer (AHPbc), and Neosealer Flo (Neo). Standardized discs and 1:1, 1:2, and 1:4 eluates of the tested materials were prepared. The following in vitro experiments were carried out: ion release, cell metabolic activity 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell migration, immunofluorescence experiment, cell attachment, gene expression, and mineralization assay. Statistical analyses were performed using one-way ANOVA followed by Tukey's post hoc test (p < .05). Increased Ca2+ release was detected in TFbc compared to AHPbc and Neo (*p < .05). Biological assays showed a discrete cell metabolic activity and cell migration in Neo-treated cell, whereas scanning electronic microscopy assay exhibited that TFbc group had a better cell adhesion process of substrate attachment, spreading, and cytoskeleton development on the niche-like structures of the cement than AHPbc and Neo. The sealers tested were able to induce overexpression of the CEMP-1, ALP, and COL1A1 genes in the first days of exposure, particularly in the case of TFbc (***p < .001). All materials tested significantly increased the mineralization of hPDLSCs when compared to the negative control, although more pronounced calcium deposition was observed in the TFbc-treated cells (***p < .001). Our results suggested that TFbc promotes cell differentiation, both by increasing the expression of key osteo/odontogenic genes and by promoting mineralization of the extracellular matrix, whereas this phenomenon was less evident in Neo and AHPbc. RESEARCH HIGHLIGHTS: TFbc group had a better cell adhesion process of substrate attachment, spreading, and cytoskeleton development on the niche-like structures of the cement than AHPbc and Neo. The sealers tested were able to induce overexpression of the CEMP-1, ALP, and COL1A1 genes in the first days of exposure, particularly in the case of TFbc. All materials tested significantly increased the mineralization of hPDLSCs when compared to the negative control, although more pronounced calcium deposition was observed in the TFbc-treated cells.


Asunto(s)
Compuestos de Calcio , Diferenciación Celular , Cerámica , Osteogénesis , Ligamento Periodontal , Silicatos , Células Madre , Humanos , Ligamento Periodontal/citología , Ligamento Periodontal/efectos de los fármacos , Compuestos de Calcio/farmacología , Compuestos de Calcio/química , Silicatos/farmacología , Silicatos/química , Diferenciación Celular/efectos de los fármacos , Cerámica/química , Células Madre/efectos de los fármacos , Células Madre/citología , Osteogénesis/efectos de los fármacos , Células Cultivadas , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cementogénesis/efectos de los fármacos , Microscopía Electrónica de Rastreo
3.
Biochem Biophys Res Commun ; 587: 9-15, 2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-34861472

RESUMEN

OBJECTIVE: The role of circadian clock in cementogenesis is unclear. This study examines the role of REV-ERBs, one of circadian clock proteins, in proliferation, migration and mineralization of cementoblasts to fill the gap in knowledge. METHODS: Expression pattern of REV-ERBα in cementoblasts was investigated in vivo and in vitro. CCK-8 assay, scratch wound healing assay, alkaline phosphatase (ALP) and alizarin red S (ARS) staining were performed to evaluate the effects of REV-ERBs activation by SR9009 on proliferation, migration and mineralization of OCCM-30, an immortalized cementoblast cell line. Furthermore, mineralization related markers including osterix (OSX), ALP, bone sialoprotein (BSP) and osteocalcin (OCN) were evaluated. RESULTS: Strong expression of REV-ERBα was found in cellular cementum around tooth apex. Rev-erbα mRNA oscillated periodically in OCCM-30 and declined after mineralization induction. REV-ERBs activation by SR9009 inhibited proliferation but promoted migration of OCCM-30 in vitro. Results of ALP and ARS staining suggested that REV-ERBs activation negatively regulated mineralization of OCCM-30. Mechanically, REV-ERBs activation attenuated the expression of OSX and its downstream targets including ALP, BSP and OCN. CONCLUSIONS: REV-ERBs are involved in cementogenesis and negatively regulate mineralization of cementoblasts via inhibiting OSX expression. Our study provides a potential target regarding periodontal and cementum regeneration.


Asunto(s)
Relojes Biológicos/genética , Calcificación Fisiológica/genética , Cemento Dental/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Cementogénesis/efectos de los fármacos , Cementogénesis/genética , Cemento Dental/citología , Cemento Dental/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Humanos , Sialoproteína de Unión a Integrina/genética , Sialoproteína de Unión a Integrina/metabolismo , Ratones , Ratones Endogámicos C57BL , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Pirrolidinas/farmacología , Transducción de Señal , Factor de Transcripción Sp7/genética , Factor de Transcripción Sp7/metabolismo , Tiofenos/farmacología
4.
Int J Mol Sci ; 22(10)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067898

RESUMEN

The aim of the study was to examine the efficacy of cold atmospheric plasma (CAP) on the mineralization and cell proliferation of murine dental cementoblasts. Cells were treated with CAP and enamel matrix derivates (EMD). Gene expression of alkaline phosphatase (ALP), bone gamma-carboxyglutamate protein (BGLAP), periostin (POSTN), osteopontin (OPN), osterix (OSX), collagen type I alpha 1 chain (COL1A1), dentin matrix acidic phosphoprotein (DMP)1, RUNX family transcription factor (RUNX)2, and marker of proliferation Ki-67 (KI67) was quantified by real-time PCR. Protein expression was analyzed by immunocytochemistry and ELISA. ALP activity was determined by ALP assay. Von Kossa and alizarin red staining were used to display mineralization. Cell viability was analyzed by XTT assay, and morphological characterization was performed by DAPI/phalloidin staining. Cell migration was quantified with an established scratch assay. CAP and EMD upregulated both mRNA and protein synthesis of ALP, POSTN, and OPN. Additionally, DMP1 and COL1A1 were upregulated at both gene and protein levels. In addition to upregulated RUNX2 mRNA levels, treated cells mineralized more intensively. Moreover, CAP treatment resulted in an upregulation of KI67, higher cell viability, and improved cell migration. Our study shows that CAP appears to have stimulatory effects on regeneration-associated cell functions in cementoblasts.


Asunto(s)
Cementogénesis/efectos de los fármacos , Cemento Dental/metabolismo , Gases em Plasma/farmacología , Animales , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ratones , Osteocalcina/metabolismo , Osteopontina/metabolismo , Gases em Plasma/metabolismo , Transcriptoma/genética
5.
J Cell Physiol ; 236(3): 2070-2086, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32740946

RESUMEN

Intermittent parathyroid hormone (PTH) promotes periodontal repair, but the underlying mechanisms remained unclear. Recent studies found that ephrinB2-EPHB4 forward signaling mediated the anabolic effect of PTH in bone homeostasis. Considering the similarities between cementum and bone, we aimed to examine the therapeutic effect of PTH on resorbed roots and explore the role of forward signaling in this process. In vivo experiments showed that intermittent PTH significantly accelerated the regeneration of root resorption and promoted expression of EPHB4 and ephrinB2. When the signaling was blocked, the resorption repair was also delayed. In vitro studies showed that intermittent PTH promoted the expression of EPHB4 and ephrinB2 in OCCM-30 cells. The effects of PTH on the mineralization capacity of OCCM-30 cells was mediated through the ephrinB2-EPHB4 forward signaling. These results support the premise that the anabolic effects of intermittent PTH on the regeneration of root resorption is via the ephrinB2-EPHB4 forward signaling pathway.


Asunto(s)
Cementogénesis/efectos de los fármacos , Efrina-B2/metabolismo , Hormona Paratiroidea/farmacología , Receptor EphB4/metabolismo , Transducción de Señal , Animales , Línea Celular , Cemento Dental/efectos de los fármacos , Cemento Dental/metabolismo , Masculino , Ratones , Modelos Biológicos , Hormona Paratiroidea/administración & dosificación , Ratas Wistar , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tomografía Computarizada por Rayos X , Raíz del Diente/diagnóstico por imagen , Raíz del Diente/efectos de los fármacos
6.
J Cell Mol Med ; 24(14): 7939-7948, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32510818

RESUMEN

Cementum regeneration, as one of the most difficult challenges of periodontal regeneration, is influenced by inflammatory factors. Inflammation may hamper or promote periodontal tissue repair under different circumstances, as it is found to do in dentin-pulp complex and bone tissue. Our team demonstrated that YAP promotes mineralization of OCCM, a cementoblast cell line. However, the effect of YAP on its mineralization under inflammatory microenvironment is unclear. In this study, cementogenesis in vitro was up-regulated after transient TNF-α treatment for 30 minutes. YAP expression also was increased by TNF-α treatment. YAP overexpression promoted OCCM mineralization after the cells were transiently treated with TNF-α because YAP overexpression inhibited NF-κB pathway activity, while YAP knockdown elevated it. The inhibited mineralization potential and activated NF-κB pathway activity by YAP knockdown also were partly rescued by the application of the NF-κB inhibitor Bay 11-7082. These results demonstrated that YAP plays a positive role in the mineralization of TNF-α transiently treated cementoblast, partly by inhibiting the NF-κB pathway activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Cementogénesis , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Cementogénesis/efectos de los fármacos , Citocinas/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Técnicas de Silenciamiento del Gen , Mediadores de Inflamación/metabolismo , Ratones , Proteínas Señalizadoras YAP
7.
Connect Tissue Res ; 60(5): 444-451, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30897979

RESUMEN

Aim: The purpose of this study was to investigate the influence of mechanical strain on OCCM-30 cementoblast differentiation and Wnt/ß-catenin pathway activity. Materials and Methods: Mechanical tension in the form of 2500-µ strain was applied to the cells using the Forcel four-point bending system, with or without the Wnt signaling activator, lithium chloride. Changes in cell differentiation and the expression of Wnt/ß-catenin pathway components in response to strain and lithium chloride were assessed by real-time PCR, immunofluorescence, and western blotting. Results: The mRNA expression levels of the cementoblastogenesis-related genes alkaline phosphatase, runt-related transcription factor 2, and collagen 1, were decreased with mechanical strain. Similarly, the Wnt signaling pathway component genes LRP5, AXIN2, and LEF1 were decreased. The immunofluorescence assay demonstrated that scant ß-catenin underwent nuclear translocation after the cells were subjected to mechanical strain. Moreover, western blotting showed that the protein levels of both ß-catenin and phosphorylated ß-catenin were increased after mechanical strain. In the presence of lithium chloride, the differentiation that was suppressed by mechanical strain was attenuated. Conclusions: 2500-µ strain mechanical strain inhibited cementoblast differentiation activity in vitro, which could be alleviated by actviating Wnt/ß-catenin signaling using lithium chloride.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cemento Dental/citología , Cloruro de Litio/farmacología , Estrés Mecánico , Animales , Línea Celular , Cementogénesis/efectos de los fármacos , Cementogénesis/genética , Cemento Dental/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , beta Catenina/metabolismo
8.
Connect Tissue Res ; 60(5): 419-430, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30734591

RESUMEN

Introduction: Preameloblast-conditioned medium (PA-CM), as a mixture of dental epithelium-derived factors, has been reported to regenerate dentin and periodontal tissues in vitro and in vivo. The aim of this study was to investigate the biological effect of Cpne7 on the proliferation, migration, and cementoblast differentiation of periodontal cells in vitro, and on the regeneration of periodontal tissue using periodontal defect model with canine in vivo. Materials and methods: The effect of Cpne7 on cell proliferation, migration, and cementoblast differentiation of periodontal cells were evaluated in vitro. A periodontal defect canine model was designed and the defects were divided into five groups: Group 1: No treatment (negative control), Group 2: Collagen carrier only, Group 3: PA-CM with collagen carrier (positive control), Group 4: PA-CM + CPNE7 Antibody (Ab) with collagen carrier, and Group 5: recombinant CPNE7 (rCPNE7) protein with collagen carrier. Results: Cpne7 was expressed in HERS cells and periodontal ligament (PDL) fibers. By real-time PCR, Cpne7 increased expression of Cap compared to the control. In the periodontal defect canine model, rCPNE7 or PA-CM regenerated periodontal complex, and the arrangement of the newly formed PDL-like fibers were perpendicular to the newly formed cementum and alveolar bone like Sharpey's fibers in natural teeth, while PA-CM + CPNE7 Ab showed irregular arrangement of the newly formed PDL-like fibers compared to the rCPNE7 or PA-CM group. Conclusion: These findings suggest that Cpne7 may have a functional role in periodontal regeneration by supporting periodontal cell attachment to cementum and facilitating physiological arrangement of PDL fibers.


Asunto(s)
Proteínas de la Membrana/metabolismo , Periodoncio/fisiología , Regeneración , Adolescente , Ameloblastos/citología , Ameloblastos/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cementogénesis/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Perros , Humanos , Ratones , Periodoncio/citología , Proteínas Recombinantes/farmacología , Regeneración/efectos de los fármacos , Diente/crecimiento & desarrollo , Diente/metabolismo , Adulto Joven
9.
Bone ; 105: 134-147, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28866368

RESUMEN

Although acellular cementum is essential for tooth attachment, factors directing its development and regeneration remain poorly understood. Inorganic pyrophosphate (PPi), a mineralization inhibitor, is a key regulator of cementum formation: tissue-nonspecific alkaline phosphatase (Alpl/TNAP) null mice (increased PPi) feature deficient cementum, while progressive ankylosis protein (Ank/ANK) null mice (decreased PPi) feature increased cementum. Bone sialoprotein (Bsp/BSP) and osteopontin (Spp1/OPN) are multifunctional extracellular matrix components of cementum proposed to have direct and indirect effects on cell activities and mineralization. Studies on dentoalveolar development of Bsp knockout (Bsp-/-) mice revealed severely reduced acellular cementum, however underlying mechanisms remain unclear. The similarity in defective cementum phenotypes between Bsp-/- mice and Alpl-/- mice (the latter featuring elevated PPi and OPN), prompted us to examine whether BSP is operating by modulating PPi-associated genes. Genetic ablation of Bsp caused a 2-fold increase in circulating PPi, altered mRNA expression of Alpl, Spp1, and Ank, and increased OPN protein in the periodontia. Generation of a Bsp knock-out (KO) cementoblast cell line revealed significantly decreased mineralization capacity, 50% increased PPi in culture media, and increased Spp1 and Ank mRNA expression. While addition of 2µg/ml recombinant BSP altered Spp1, Ank, and Enpp1 expression in cementoblasts, changes resulting from this dose were not dependent on the integrin-binding RGD motif or MAPK/ERK signaling pathway. Decreasing PPi by genetic ablation of Ank on the Bsp-/- mouse background reestablished cementum formation, allowing >3-fold increased acellular cementum volume compared to wild-type (WT). However, deleting Ank did not fully compensate for the absence of BSP. Bsp-/-; Ank-/- double-deficient mice exhibited mean 20-27% reduced cementum thickness and volume compared to Ank-/- mice. From these data, we conclude that the perturbations in PPi metabolism are not solely driving the cementum pathology in Bsp-/- mice, and that PPi is more potent than BSP as a cementum regulator, as shown by the ability to override loss of BSP by lowering PPi. We propose that BSP and PPi work in concert to direct mineralization in cementum and likely other mineralized tissues.


Asunto(s)
Calcificación Fisiológica , Cementogénesis/efectos de los fármacos , Difosfatos/farmacología , Sialoproteína de Unión a Integrina/metabolismo , Animales , Calcificación Fisiológica/efectos de los fármacos , Cemento Dental/efectos de los fármacos , Cemento Dental/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Sialoproteína de Unión a Integrina/deficiencia , Ratones Noqueados , Periodoncio/metabolismo , Fenotipo , Proteínas de Transporte de Fosfato/metabolismo , Fosforilación/efectos de los fármacos
10.
Acta Biomater ; 61: 134-143, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28764948

RESUMEN

Cementum formation on the exposed tooth-root surface is a critical process in periodontal regeneration. Although various therapeutic approaches have been developed, regeneration of integrated and functional periodontal complexes is still wanting. Here, we found that the OCCM30 cementoblasts cultured on fibrin matrix express substantial levels of matrix proteinases, leading to the degradation of fibrin and the apoptosis of OCCM30 cells, which was reversed upon treatment with a proteinase inhibitor, ε-aminocaproic acid (ACA). Based on these findings, ACA-releasing chitosan particles (ACP) were fabricated and ACP-incorporated fibrin (fibrin-ACP) promoted the differentiation of cementoblasts in vitro, as confirmed by bio-mineralization and expressions of molecules associated with mineralization. In a periodontal defect model of beagle dogs, fibrin-ACP resulted in substantial cementum formation on the exposed root dentin in vivo, compared to fibrin-only and enamel matrix derivative (EMD) which is used clinically for periodontal regeneration. Remarkably, the fibrin-ACP developed structural integrations of the cementum-periodontal ligament-bone complex by the Sharpey's fiber insertion. In addition, fibrin-ACP promoted alveolar bone regeneration through increased bone volume of tooth roof-of-furcation defects and root coverage. Therefore, fibrin-ACP can promote cementogenesis and osteogenesis by controlling biodegradability of fibrin, implicating the feasibility of its therapeutic use to improve periodontal regeneration. STATEMENT OF SIGNIFICANCE: Cementum, the mineralized layer on root dentin surfaces, functions to anchor fibrous connective tissues on tooth-root surfaces with the collagenous Sharpey's fibers integration, of which are essential for periodontal functioning restoration in the complex. Through the cementum-responsible fiber insertions on tooth-root surfaces, PDLs transmit various mechanical responses to periodontal complexes against masticatory/occlusal stimulations to support teeth. In this study, periodontal tissue regeneration was enhanced by use of modified fibrin biomaterial which significantly promoted cementogenesis within the periodontal complex with structural integration by collagenous Sharpey's fiber insertions in vivo by controlling fibrin degradation and consequent cementoblast apoptosis. Furthermore, the modified fibrin could improve repair and regeneration of tooth roof-of-furcation defects, which has spatial curvatures and geometrical difficulties and hardly regenerates periodontal tissues.


Asunto(s)
Ácido Aminocaproico/química , Diferenciación Celular/efectos de los fármacos , Quitosano/química , Cemento Dental/citología , Fibrina/farmacología , Regeneración , Animales , Apoptosis/efectos de los fármacos , Bovinos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cementogénesis/efectos de los fármacos , Cemento Dental/diagnóstico por imagen , Cemento Dental/efectos de los fármacos , Perros , Masculino , Ratones , Nanopartículas/química , Periodoncio/diagnóstico por imagen , Periodoncio/efectos de los fármacos , Periodoncio/fisiología , Ratas , Regeneración/efectos de los fármacos , Microtomografía por Rayos X
11.
Mol Cells ; 40(8): 550-557, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28835018

RESUMEN

The periodontal ligament (PDL) is the connective tissue between tooth root and alveolar bone containing mesenchymal stem cells (MSC). It has been suggested that human periodontal ligament stem cells (hPDLSCs) differentiate into osteo/cementoblast and ligament progenitor cells. The periodontitis is a representative oral disease where the PDL tissue is collapsed, and regeneration of this tissue is important in periodontitis therapy. Fibroblast growth factor-2 (FGF-2) stimulates proliferation and differentiation of fibroblastic MSCs into various cell lineages. We evaluated the dose efficacy of FGF-2 for cytodifferentiation of hPDLSCs into ligament progenitor. The fibrous morphology was highly stimulated even at low FGF-2 concentrations, and the expression of teno/ligamentogenic markers, scleraxis and tenomodulin in hPDLSCs increased in a dose dependent manner of FGF-2. In contrast, expression of the osteo/cementogenic markers decreased, suggesting that FGF-2 might induce and maintain the ligamentogenic potential of hPDLSCs. Although the stimulation of tenocytic maturation by TGF-ß1 was diminished by FGF-2, the inhibition of the expression of early ligamentogenic marker by TGF-ß1 was redeemed by FGF-2 treatment. The stimulating effect of BMPs on osteo/cementogenesis was apparently suppressed by FGF-2. These results indicate that FGF-2 predominantly differentiates the hPDLSCs into teno/ligamentogenesis, and has an antagonistic effect on the hard tissue differentiation induced by BMP-2 and BMP-4.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Cementogénesis/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Osteogénesis/efectos de los fármacos , Ligamento Periodontal/citología , Células Madre/citología , Tendones/citología , Factor de Crecimiento Transformador beta1/farmacología , Adulto , Biomarcadores/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Cementogénesis/genética , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Osteogénesis/genética , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Adulto Joven
12.
BMC Cell Biol ; 18(1): 19, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28427342

RESUMEN

BACKGROUND: External root resorption, commonly starting from cementum, is a severe side effect of orthodontic treatment. In this pathological process and repairing course followed, cementoblasts play a significant role. Previous studies implicated that parathyroid hormone (PTH) could act on committed osteoblast precursors to promote differentiation, and inhibit apoptosis. But little was known about the role of PTH in cementoblasts. The purpose of this study was to investigate the effects of intermittent PTH on cementoblasts and its influence after mechanical strain treatment. RESULTS: Higher levels of cementogenesis- and differentiation-related biomarkers (bone sialoprotein (BSP), osteocalcin (OCN), Collagen type I (COL1) and Osterix (Osx)) were shown in 1-3 cycles of intermittent PTH treated groups than the control group. Additionally, intermittent PTH increased alkaline phosphatase (ALP) activity and mineralized nodules formation, as measured by ALP staining, quantitative ALP assay, Alizarin red S staining and quantitative calcium assay. The morphology of OCCM-30 cells changed after mechanical strain exertion. Expression of BSP, ALP, OCN, osteopontin (OPN) and Osx was restrained after 18 h mechanical strain. Furthermore, intermittent PTH significantly increased the expression of cementogenesis- and differentiation-related biomarkers in mechanical strain treated OCCM-30 cells. CONCLUSIONS: Taken together, these data suggested that intermittent PTH promoted cementum formation through activating cementogenesis- and differentiation-related biomarkers, and attenuated the catabolic effects of mechanical strain in immortalized cementoblasts OCCM-30.


Asunto(s)
Cementogénesis/efectos de los fármacos , Cemento Dental/citología , Cemento Dental/efectos de los fármacos , Hormona Paratiroidea/farmacología , Estrés Mecánico , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Cementogénesis/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cemento Dental/enzimología , Regulación de la Expresión Génica/efectos de los fármacos , Sialoproteína de Unión a Integrina/genética , Sialoproteína de Unión a Integrina/metabolismo , Ratones , Osteocalcina/genética , Osteocalcina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , Hormona Paratiroidea/administración & dosificación , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Factor de Transcripción Sp7/genética , Factor de Transcripción Sp7/metabolismo , Factores de Tiempo , Raíz del Diente/citología , Raíz del Diente/efectos de los fármacos
13.
Mater Sci Eng C Mater Biol Appl ; 73: 726-735, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28183667

RESUMEN

Recent exciting findings of the biological interactions of graphene materials have shed light on potential biomedical applications of graphene-containing composites. Owing to the superior mechanical properties and low coefficient of thermal expansion, graphene has been widely used in the reinforcement of biocomposites. In the present study, various ratios of graphene (0.25wt%, 0.5wt% and 1.0wt%) were reinforced into calcium silicate (CS) for bone graft application. Results show that the graphene was embedded in the composites homogeneously. Adding 1wt% graphene into CS increased the young's modulus by ~47.1%. The formation of bone-like apatite on a range of composites with graphene weight percentages ranging from 0 to 1 has been investigated in simulated body fluid. The presence of a bone-like apatite layer on the composites surface after immersion in simulated body fluid was considered by scanning electron microscopy. In vitro cytocompatibility of the graphene-contained CS composites was evaluated using human marrow stem cells (hMSCs). The proliferation and alkaline phosphatase, osteopontin and osteocalcin osteogenesis-related protein expression of the hMSCs on the 1wt% graphene-contained specimens showed better results than on the pure CS. In addition, the angiogenesis-related protein (vWF and ang-1) secretion of cells was significantly stimulated when the graphene concentration in the composites was increased. These results suggest that graphene-contained CS bone graft are promising materials for bone tissue engineering applications.


Asunto(s)
Materiales Biocompatibles/farmacología , Compuestos de Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Grafito/farmacología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Silicatos/farmacología , Fosfatasa Alcalina/metabolismo , Western Blotting , Cementos para Huesos/farmacología , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cementogénesis/efectos de los fármacos , Colágeno Tipo I/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Peso Molecular , Osteocalcina/metabolismo , Espectroscopía de Fotoelectrones , Resistencia a la Tracción , Difracción de Rayos X
14.
Int J Nanomedicine ; 11: 3145-58, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27471382

RESUMEN

Periodontitis is a major cause for tooth loss, which affects about 15% of the adult population. Cementum regeneration has been the crux of constructing the periodontal complex. Cementum protein 1 (CEMP1) is a cementum-specific protein that can induce cementogenic differentiation. In this study, poly(ethylene glycol) (PEG)-stabilized amorphous calcium phosphate (ACP) nanoparticles were prepared by wet-chemical method and then loaded with recombinant human CEMP1 (rhCEMP1) for controlled release. An electrospun multiphasic scaffold constituted of poly(ε-caprolactone) (PCL), type I collagen (COL), and rhCEMP1/ACP was fabricated. The effects of rhCEMP1/ACP/PCL/COL scaffold on the attachment proliferation, osteogenic, and cementogenic differentiations of human periodontal ligament cells, (PDLCs) were systematically investigated. A critical size defect rat model was introduced to evaluate the effect of tissue regeneration of the scaffolds in vivo. The results showed that PEG-stabilized ACP nanoparticles formed a core-shell structure with sustained release of rhCEMP1 for up to 4 weeks. rhCEMP1/ACP/PCL/COL scaffold could suppress PDLCs proliferation behavior and upregulate the expression of cementoblastic markers including CEMP1 and cementum attachment protein while downregulating osteoblastic markers including osteocalcin and osteopontin when it was cocultured with PDLCs in vitro for 7 days. Histology analysis of cementum after being implanted with the scaffold in rats for 8 weeks showed that there was cementum-like tissue formation but little bone formation. These results indicated the potential of using electrospun multiphasic scaffolds for controlled release of rhCEMP1 for promoting cementum regeneration in reconstruction of the periodontal complex.


Asunto(s)
Cemento Dental/fisiología , Proteínas/farmacología , Proteínas Recombinantes/farmacología , Regeneración/efectos de los fármacos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Adolescente , Animales , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Fosfatos de Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cementogénesis/efectos de los fármacos , Colágeno Tipo I/farmacología , Preparaciones de Acción Retardada/farmacología , Cemento Dental/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Osteogénesis/efectos de los fármacos , Ligamento Periodontal/citología , Ligamento Periodontal/efectos de los fármacos , Poliésteres/farmacología , Ratas Wistar , Espectroscopía Infrarroja por Transformada de Fourier
15.
J Periodontal Res ; 51(6): 699-715, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26833268

RESUMEN

The molecular bases of periodontal tissue induction and regeneration are the osteogenic proteins of the transforming growth factor-ß (TGF-ß) supergene family. These morphogens act as soluble mediators for the induction of tissues morphogenesis sculpting the multicellular mineralized structures of the periodontal tissues with functionally oriented ligament fibers into newly formed cementum. Human TGF-ß3 (hTGF-ß3 ) in growth factor-reduced Matrigel® matrix induces cementogenesis when implanted in class II mandibular furcation defects surgically prepared in the non-human primate Chacma baboon, Papio ursinus. The newly formed periodontal ligament space is characterized by running fibers tightly attached to the cementoid surface penetrating as mineralized constructs within the newly formed cementum assembling and initiating within the mineralized dentine. Angiogenesis heralds the newly formed periodontal ligament space, and newly sprouting capillaries are lined by cellular elements with condensed chromatin interpreted as angioblasts responsible for the rapid and sustained induction of angiogenesis. The inductive activity of hTGF-ß3 in Matrigel® matrix is enhanced by the addition of autogenous morcellated fragments of the rectus abdominis muscle potentially providing myoblastic, pericytic/perivascular stem cells for continuous tissue induction and morphogenesis. The striated rectus abdominis muscle is endowed with stem cell niches in para/perivascular location, which can be dominant, thus imposing stem cell features or stemness to the surrounding cells. This capacity to impose stemness is morphologically shown by greater alveolar bone induction and cementogenesis when hTGF-ß3 in Matrigel® matrix is combined with morcellated fragments of autogenous rectus abdominis muscle. The induction of periodontal tissue morphogenesis develops as a mosaic structure in which the osteogenic proteins of the TGF-ß supergene family singly, synergistically and synchronously initiate and maintain tissue induction and morphogenesis. In primates, the presence of several homologous yet molecularly different isoforms with osteogenic activity highlights the biological significance of this apparent redundancy and indicates multiple interactions during embryonic development and bone regeneration in postnatal life. Molecular redundancy with associated different biological functionalities in primate tissues may simply represent the fine-tuning of speciation-related molecular evolution in anthropoid apes at the early Pliocene boundary, which resulted in finer tuning of the bone induction cascade.


Asunto(s)
Periodoncio/fisiología , Regeneración/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Cementogénesis/efectos de los fármacos , Colágeno/farmacología , Combinación de Medicamentos , Laminina/farmacología , Papio ursinus , Periodoncio/efectos de los fármacos , Primates , Proteoglicanos/farmacología , Regeneración/fisiología , Factor de Crecimiento Transformador beta/genética
16.
Biomater Sci ; 4(4): 639-51, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26806408

RESUMEN

Gingival recession is a common clinical problem that results in esthetic deficiencies and poor plaque control and predominantly occurs in aged patients. In order to restore the cervical region, ideal biomaterials should possess the ability to stimulate proliferation and osteogenesis/cementogenesis of human gingival fibroblasts (HGF) and have a strong antibiofilm effect. The aim of the present study was to investigate the interactions of HGF and oral multispecies biofilms with Ca, Mg and Si-containing bredigite (BRT, Ca7MgSi4O16) bioceramics. BRT extract induced osteogenic/cementogenic differentiation of HGF and its inhibition of plaque biofilm formation were systematically studied. BRT extract in concentrations lower than <200 mg mL(-1) presented high biocompatibility to HGF cells in 3 days. Ion extracts from BRT also stimulated a series of bone-related gene and protein expressions in HGF cells. Furthermore, BRT extract significantly inhibited oral multispecies plaque biofilm growth on its surface and contributed to over 30% bacterial cell death without additional antibacterial agents in two weeks. A planktonic killing test showed that BRT suppressed 98% plaque bacterial growth compared to blank control in 3 days. The results also revealed that BRT extract has an osteostimulation effect on HGF. The suppression effect on plaque biofilms suggested that BRT might be used as a bioactive material for cervical restoration and that the synergistic effect of bioactive ions, such as Ca, Mg and Si ions, played an important role in the design and construction of bifunctional biomaterials in combination with tissue regeneration and antibiofilm activity.


Asunto(s)
Materiales Biocompatibles/química , Biopelículas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Placa Dental/química , Placa Dental/microbiología , Fibroblastos/química , Encía/metabolismo , Iones/química , Iones/farmacología , Osteogénesis/efectos de los fármacos , Fosfatasa Alcalina/química , Fosfatasa Alcalina/metabolismo , Materiales Biocompatibles/farmacología , Diferenciación Celular/fisiología , Cementogénesis/efectos de los fármacos , Cementogénesis/fisiología , Fibroblastos/microbiología , Encía/química , Humanos , Osteogénesis/fisiología
17.
J Periodontal Res ; 51(3): 284-94, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26257338

RESUMEN

BACKGROUND AND OBJECTIVE: The predictability of conventional periodontal treatments for damaged periodontal tissue is limited, particularly on the regeneration of new cementum. As signaling molecules, a range of growth factors has been used to promote periodontal regeneration on periodontal ligament (PDL) and cementum defects. A preameloblast-conditioned medium (PA-CM) was prepared from cultured murine apical bud cells, which can differentiate into ameloblasts. We examined the effect of PA-CM on PDL cells and cementoblasts in vitro and evaluated histologically the effects of PA-CM on the regeneration of experimentally induced periodontal defects in vivo. MATERIAL AND METHODS: In vitro, the effects of PA-CM on the migration of human PDL cells were examined using a scratch wound healing assay and a transwell assay. The differentiation and mineralization potential of PA-CM-treated human PDL cells and murine cementoblastic OCCM-30 cells was examined by real-time polymerase chain reaction and Alizarin red-S staining. In vivo, six mongrel dogs (12-16 kg; 6-8 mo old) were used. Twenty-four roots were replanted with either, (i) only periodontal defects (n = 12; control group), or (ii) periodontal defects and PA-CM treatment (n = 12; experimental group). In the experimental group, the PDL and cementum between notches was removed using a Gracey curette and soaked in 0.08 mL water containing 80 µg of a PA-CM for 2 min. The dogs were killed at 4 and 8 wk post-surgery. RESULTS: The in vitro results showed that PA-CM stimulated the migration of PDL cells and promoted the differentiation and mineralization of PDL cells and cementoblasts. Real-time polymerase chain reaction analysis revealed stronger expression of Runx2, Osx, OC, Bsp and Cap mRNAs in the PA-CM-treated PDL cells and cementoblasts than those in the control cells. In vivo, newly formed PDL-like tissue and cementum-like tissue were observed partially between the root surfaces and newly formed bone in the experimental group. The regenerated PDL-like tissue in the experimental group was significantly higher than that in the control group at 8 wk (p < 0.05). The replacement resorption on the experimental group was significantly lower than that in the control group at 8 wk (p < 0.05). In addition, the amount of newly formed cementum-like tissue in the experimental group was significantly higher than that in the control group at 4 and 8 wk (p < 0.05). CONCLUSION: These results suggest that PA-CM has the potential to regenerate periodontal tissues in PDL and cementum defects.


Asunto(s)
Cemento Dental/efectos de los fármacos , Ligamento Periodontal/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Adolescente , Animales , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ensayos de Migración Celular , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Cementogénesis/efectos de los fármacos , Medios de Cultivo Condicionados , Cemento Dental/lesiones , Perros , Humanos , Ratones Endogámicos C57BL , Tercer Molar , Periodoncio/efectos de los fármacos , Periodoncio/lesiones , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Raíz del Diente/efectos de los fármacos , Raíz del Diente/lesiones , Raíz del Diente/patología , Adulto Joven
18.
Int J Nanomedicine ; 10: 7031-44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26648716

RESUMEN

The surface structure of bioceramic scaffolds is crucial for its bioactivity and osteoinductive ability, and in recent years, human periodontal ligament stem cells have been certified to possess high osteogenic and cementogenic differential ability. In the present study, hydroxyapatite (HA) bioceramics with micro-nano-hybrid surface (mnHA [the hybrid of nanorods and microrods]) were fabricated via hydrothermal reaction of the α-tricalcium phosphate granules as precursors in aqueous solution, and the effects of mnHA on the attachment, proliferation, osteogenic and cementogenic differentiations of human periodontal ligament stem cells as well as the related mechanisms were systematically investigated. The results showed that mnHA bioceramics could promote cell adhesion, proliferation, alkaline phosphatase (ALP) activity, and expression of osteogenic/cementogenic-related markers including runt-related transcription factor 2 (Runx2), ALP, osteocalcin (OCN), cementum attachment protein (CAP), and cementum protein (CEMP) as compared to the HA bioceramics with flat and dense surface. Moreover, mnHA bioceramics stimulated gene expression of low-density lipoprotein receptor-related protein 5 (LRP5) and ß-catenin, which are the key genes of canonical Wnt signaling. Moreover, the stimulatory effect on ALP activity and osteogenic and cementogenic gene expression, including that of ALP, OCN, CAP, CEMP, and Runx2 of mnHA bioceramics could be repressed by canonical Wnt signaling inhibitor dickkopf1 (Dkk1). The results suggested that the HA bioceramics with mnHA could act as promising grafts for periodontal tissue regeneration.


Asunto(s)
Cementogénesis/efectos de los fármacos , Cerámica/farmacología , Durapatita/farmacología , Nanopartículas/química , Osteogénesis/efectos de los fármacos , Ligamento Periodontal/citología , Células Madre/citología , Vía de Señalización Wnt/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Madre/efectos de los fármacos , Células Madre/ultraestructura
19.
Tissue Eng Part A ; 21(23-24): 2817-28, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25808697

RESUMEN

The periodontium, consisting of gingiva, periodontal ligament (PDL), cementum, and alveolar bone, is necessary for the maintenance of tooth function. Specifically, the regenerative abilities of cementum with inserted PDL are important for the prevention of tooth loss. Periodontal ligament stem cells (PDLSCs), which are located in the connective tissue PDL between the cementum and alveolar bone, are an attractive candidate for hard tissue formation. We investigated the effects of recombinant human plasminogen activator inhibitor-1 (rhPAI-1) on cementogenic differentiation of human PDLSCs (hPDLSCs) in vitro and in vivo. Untreated and rhPAI-1-treated hPDLSCs mixed with hydroxyapatite/tricalcium phosphate (HA/TCP) and dentin matrix were transplanted subcutaneously into the dorsal surface of immunocompromised mice to assess their capacity for hard tissue formation at 8 and 10 weeks posttransplantation. rhPAI-1 accelerated mineral nodule formation and increased the mRNA expression of cementoblast-associated markers in hPDLSCs. We also observed that rhPAI-1 upregulated the levels of osterix (OSX) and cementum protein 1 (CEMP1) through Smad2/3 and p38 pathways, whereas specific inhibitors of Smad3 and p38 inhibited the enhancement of mineralization of hPDLSCs by rhPAI-1. Furthermore, transplantation of hPDLSCs with rhPAI-1 showed a great ability to promote cementogenic differentiation. Notably, rhPAI-1 induced hPDLSCs to regenerate cementum-like tissue with PDL fibers inserted into newly formed cementum-like tissue. These results suggest that rhPAI-1 may play a key role in cementogenic differentiation of hPDLSCs. rhPAI-1 with hPDLSCs may be a good candidate for future clinical applications in periodontal tissue regeneration and possibly in tooth root bioengineering.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cementogénesis/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/farmacología , Células Madre/metabolismo , Adolescente , Adulto , Animales , Células Cultivadas , Femenino , Historia Antigua , Humanos , Masculino , Ratones , Ligamento Periodontal , Proteínas Recombinantes/farmacología , Células Madre/citología
20.
J Clin Periodontol ; 42(1): 62-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25361403

RESUMEN

AIM: To evaluate the effects of fibroblast growth factor (FGF)-2 on the healing of surgical periodontal defects in rats with early, streptozotocin-induced diabetes. MATERIALS AND METHODS: Fifty Wistar rats were assigned to streptozotocin-induced diabetes or non-diabetes group. Periodontal defects were surgically created at maxillary first molars. Defects were treated with hydroxypropyl cellulose (HPC) or FGF-2 with HPC. Defect fill was evaluated by microcomputed tomography. Histological and immunohistochemical analyses were performed. RESULTS: Compared to vehicle alone, FGF-2 treatment yielded significantly greater bone volume and trabecular thickness in diabetes group. Diabetes group displayed reduced new bone formation and significantly longer epithelial down-growth compared to non-diabetes group. In diabetes group, FGF-2 treatment increased PCNA-positive cells and new bone formation after 2 weeks and suppressed epithelial down-growth, but new cementum formation was minimal even after 4 weeks. In diabetes group, overexpression of vascular endothelial growth factor was evident in cells within connective tissue, and no significant enhancement was observed by FGF-2 treatment. FGF-2 increased the expression of α-smooth muscle actin in diabetes group. CONCLUSIONS: Treatment of surgical periodontal defects in diabetic rats with the single application of FGF-2 provided beneficial effects primarily on new bone formation via increasing cell proliferation and regulating angiogenesis.


Asunto(s)
Pérdida de Hueso Alveolar/tratamiento farmacológico , Diabetes Mellitus Experimental/complicaciones , Factor 2 de Crecimiento de Fibroblastos/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Actinas/análisis , Actinas/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Celulosa/análogos & derivados , Cementogénesis/efectos de los fármacos , Tejido Conectivo/efectos de los fármacos , Portadores de Fármacos , Inserción Epitelial/efectos de los fármacos , Masculino , Enfermedades Maxilares/tratamiento farmacológico , Diente Molar/patología , Osteogénesis/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/análisis , Ratas , Ratas Wistar , Estreptozocina , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Microtomografía por Rayos X/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA