Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Dent Mater J ; 42(4): 610-616, 2023 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-37438118

RESUMO

Recently, the use of orthodontic mini-screws as an anchorage for orthodontic treatment is increasing, and the degree of osseointegration of the mini-screws affects the performance of orthodontic treatment. This study aimed to evaluate the biocompatibility and osseointegration of Titanium 6Aluminum 4Vanadium (Ti-6Al-4V) alloy orthodontic mini-screws with an ibandronate-loaded TiO2 nanotube (TNT) layer. The TNT layer was formed on the surface of the Ti-6Al-4V alloy orthodontic mini-screws and loaded with ibandronate. The TNT formed by anodic oxidation formed a completely self-organized and compact structure and was stably released for 7 days after loading with ibandronate. Mini-screws loaded with ibandronate were implanted into both tibias of rats, confirming rapid initial bone regeneration. We demonstrate that the release of stable ibandronate from the TNT layer of Ti-6Al-4V alloy orthodontic mini-screws can effectively improve biocompatibility and osseointegration.


Assuntos
Implantes Dentários , Nanotubos , Ratos , Animais , Titânio/química , Osseointegração , Ácido Ibandrônico , Ligas , Parafusos Ósseos , Propriedades de Superfície
2.
Antioxidants (Basel) ; 11(12)2022 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-36552528

RESUMO

Hyperglycemia has various adverse health effects, some of which are due to chronic oxidative and inflammatory impairment of bone marrow (BM), hematopoietic stem cells (HSCs), and mesenchymal stem cells (MSCs). Astaxanthin (ASTX) has been shown to ameliorate hyperglycemia-associated systemic complications and acute mortality, and this effect is partially associated with restoration of normal hematopoiesis. Here, the effects of ASTX on diabetes-induced complications in BM and BM stem cells were investigated, and the underlying molecular mechanisms were elucidated. Ten-week-old C57BL/6 mice received a single intraperitoneal injection of streptozotocin (STZ; 150 mg/kg) in combination with oral gavage of ASTX (12.5 mg/kg) for 30 or 60 consecutive days. Supplemental ASTX ameliorated acute mortality and restored the STZ-impaired bone mass accrual and BM microenvironment in STZ-injected mice. Oral gavage of ASTX suppressed osteoclast formation in the BM of STZ-injected mice. Specifically, supplementation with ASTX inhibited oxidative stress and senescence induction of BM HSCs and MSCs and ameliorated hematopoietic disorders in STZ-injected mice. These effects of ASTX were associated with BM restoration of angiopoietin 1, stromal cell-derived factor 1, ß-catenin, and Nrf2. Long-term ASTX gavage also recovered the STZ-induced dysfunction in migration, colony formation, and mineralization of BM-derived stromal cells. Further, a direct addition of ASTX exhibited direct and dose-dependent inhibition of osteoclastic activation without cytotoxic effects. Collectively, these results indicate that ASTX protects against diabetes-induced damage in the BM microenvironment in BM, HSCs, and MSCs and restores normal hematopoiesis and bone accrual in STZ-injected mice.

3.
Mater Sci Eng C Mater Biol Appl ; 135: 112673, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35581065

RESUMO

Scaffolds combined with bioactive agents can enhance bone regeneration at therapeutic sites. We explore whether combined supplementation with coumaric acid and recombinant human-cartilage oligomeric matrix protein-angiopoietin 1 (rhCOMP-Ang1) is an ideal approach for bone tissue engineering. We developed coumaric acid-conjugated absorbable collagen scaffold (CA-ACS) and investigated whether implanting CA-ACS in combination with rhCOMP-Ang1 facilitates ACS- or CA-ACS-mediated bone formation using a rat model of critically sized mandible defects. We examined the mechanisms by which coumaric acid and rhCOMP-Ang1 regulate behaviors of human periodontal ligament fibroblasts (hPLFs). The CA-ACS exhibits greater anti-degradation and mechanical strength properties than does ACS alone. Implanting CA-ACS loaded with rhCOMP-Ang1 greatly enhances bone regeneration at the defect via the activation of angiogenic, osteogenic, and anti-osteoclastic responses compared with other rat groups implanted with an ACS alone or CA-ACS. Treatment with both rhCOMP-Ang1 and coumaric acid increases proliferation, mineralization, and migration of cultured hPLFs via activation of the Ang1/Tie2 signaling axis at a greater rate than treatment with either of them alone. Collectively, this study demonstrates that CA-ACS impregnated with rhCOMP-Ang1 enhances bone regeneration at therapeutic sites, and this enhancement is associated with a synergistic interaction between rhCOMP-Ang1-mediated angiogenesis and coumaric acid-related antioxidant responses.


Assuntos
Angiopoietina-1 , Antioxidantes , Angiopoietina-1/metabolismo , Angiopoietina-1/farmacologia , Animais , Antioxidantes/farmacologia , Proteína de Matriz Oligomérica de Cartilagem , Colágeno/farmacologia , Ácidos Cumáricos , Mandíbula , Ratos
4.
Prog Orthod ; 23(1): 11, 2022 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-35368222

RESUMO

BACKGROUND: Mini-screws are widely used as temporary anchorages in orthodontic treatment, but have the disadvantage of showing a high failure rate of about 10%. Therefore, orthodontic mini-screws should have high biocompatibility and retention. Previous studies have demonstrated that the retention of mini-screws can be improved by imparting bioactivity to the surface. The method for imparting bioactivity proposed in this paper is to sequentially perform anodization, periodic pre-calcification, and heat treatments with a Ti-6Al-4V ELI alloy mini-screw. MATERIALS AND METHODS: A TiO2 nanotube-structured layer was formed on the surface of the Ti-6Al-4V ELI alloy mini-screw through anodization in which a voltage of 20 V was applied to a glycerol solution containing 20 wt% H2O and 1.4 wt% NH4F for 60 min. Fine granular calcium phosphate precipitates of HA and octacalcium phosphate were generated as clusters on the surface through the cyclic pre-calcification and heat treatments. The cyclic pre-calcification treatment is a process of immersion in a 0.05 M NaH2PO4 solution and a saturated Ca(OH)2 solution at 90 °C for 1 min each. RESULTS: It was confirmed that the densely structured protrusions were precipitated, and Ca and P concentrations, which bind and concentrate endogenous bone morphogenetic proteins, increased on the surface after simulated body fluid (SBF) immersion test. In addition, the removal torque of the mini-screw fixed into rabbit tibias for 4 weeks was measured to be 8.70 ± 2.60 N cm. CONCLUSIONS: A noteworthy point in this paper is that the Ca and P concentrations, which provide a scaffold suitable for endogenous bone formation, further increased over time after SBF immersion of the APH group specimens. The other point is that our mini-screws have a significantly higher removal torque compared to untreated mini-screws. These results represent that the mini-screw proposed in this paper can be used as a mini-screw for orthodontics.


Assuntos
Temperatura Alta , Osseointegração , Ligas , Animais , Materiais Biocompatíveis , Parafusos Ósseos , Humanos , Coelhos , Titânio
5.
Nutrients ; 13(10)2021 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-34684576

RESUMO

Numerous studies highlight that astaxanthin (ASTX) ameliorates hyperglycemic condition and hyperglycemia-associated chronic complications. While periodontitis and periodontic tissue degradation are also triggered under chronic hyperglycemia, the roles of ASTX on diabetes-associated periodontal destruction and the related mechanisms therein are not yet fully understood. Here, we explored the impacts of supplemental ASTX on periodontal destruction and systemic complications in type I diabetic mice. To induce diabetes, C57BL/6 mice received a single intraperitoneal injection of streptozotocin (STZ; 150 mg/kg), and the hyperglycemic mice were orally administered with ASTX (12.5 mg/kg) (STZ+ASTX group) or vehicle only (STZ group) daily for 60 days. Supplemental ASTX did not improve hyperglycemic condition, but ameliorated excessive water and feed consumptions and lethality in STZ-induced diabetic mice. Compared with the non-diabetic and STZ+ASTX groups, the STZ group exhibited severe periodontal destruction. Oral gavage with ASTX inhibited osteoclastic formation and the expression of receptor activator of nuclear factor (NF)-κB ligand, 8-OHdG, γ-H2AX, cyclooxygenase 2, and interleukin-1ß in the periodontium of STZ-injected mice. Supplemental ASTX not only increased the levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and osteogenic transcription factors in the periodontium, but also recovered circulating lymphocytes and endogenous antioxidant enzyme activity in the blood of STZ-injected mice. Furthermore, the addition of ASTX blocked advanced glycation end products-induced oxidative stress and growth inhibition in human-derived periodontal ligament cells by upregulating the Nrf2 pathway. Together, our results suggest that ASTX does not directly improve hyperglycemia, but ameliorates hyperglycemia-triggered periodontal destruction and oxidative systemic complications in type I diabetes.


Assuntos
Antioxidantes/metabolismo , Diabetes Mellitus Experimental/complicações , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Periodontite/tratamento farmacológico , Periodontite/etiologia , Estreptozocina/administração & dosagem , Adolescente , Processo Alveolar/patologia , Animais , Glicemia/metabolismo , Catalase/sangue , Proliferação de Células , Citocinas/metabolismo , Dano ao DNA , Diabetes Mellitus Experimental/sangue , Suplementos Nutricionais , Comportamento Alimentar , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Hiperglicemia/complicações , Mediadores da Inflamação/metabolismo , Injeções , Linfócitos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Ligamento Periodontal/patologia , Periodontite/sangue , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/sangue , Regulação para Cima , Xantofilas/farmacologia , Xantofilas/uso terapêutico , Adulto Jovem
6.
Antioxidants (Basel) ; 10(8)2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34439457

RESUMO

While total body irradiation (TBI) is an everlasting curative therapy, the irradiation can cause long-term bone marrow (BM) injuries, along with senescence of hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) via reactive oxygen species (ROS)-induced oxidative damages. Thus, ameliorating or preventing ROS accumulation and oxidative stress is necessary for TBI-requiring clinical treatments. Here, we explored whether administration of ferulic acid, a dietary antioxidant, protects against TBI-mediated systemic damages, and examined the possible mechanisms therein. Sublethal TBI (5 Gy) decreased body growth, lifespan, and production of circulating blood cells in mice, together with ROS accumulation, and senescence induction of BM-conserved HSCs and MSCs. TBI also impaired BM microenvironment and bone mass accrual, which was accompanied by downregulated osteogenesis and by osteoclastogenic and adipogenic activation in BM. Long-term intraperitoneal injection of ferulic acid (50 mg/kg body weight, once per day for 37 consecutive days) protected mice from TBI-mediated mortality, stem cell senescence, and bone mass loss by restoring TBI-stimulated disorders in osteogenic, osteoclastic, and adipogenic activation in BM. In vitro experiments using BM stromal cells supported radioprotective effects of ferulic acid on TBI-mediated defects in proliferation and osteogenic differentiation. Overall, treatment with ferulic acid prevented TBI-mediated liver damage and enhanced endogenous antioxidant defense systems in the liver and BM. Collectively, these results support an efficient protection of TBI-mediated systemic defects by supplemental ferulic acid, indicating its clinical usefulness for TBI-required patients.

7.
Stem Cells ; 39(1): 103-114, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33038284

RESUMO

Although functional association between Wnt signaling and bone homeostasis has been well described through genetic ablation of Wntless (Wls), the mechanisms of how osteoblastic Wls regulates the fate of bone marrow stromal cells (BMSCs) and hematopoietic stem cells (HSCs) in relation to age are not yet understood. Here, we generated Col2.3-Cre;Wlsfl/fl mice that were free from premature lethality and investigated age-related impacts of osteoblastic Wls deficiency on hematopoiesis, BM microenvironment, and maintenance of BMSCs (also known as BM-derived mesenchymal stem/stromal cells) and HSCs. Ablation of osteoblastic Wls deteriorated BM microenvironment and bone mass accrual along with age-independent effects on functions of BMSCs. Osteoblastic Wls deletion impaired HSC repopulation and progeny with skewing toward myeloid lineage cells only at old stage. As proven by hallmarks of stem cell senescence, osteoblastic Wls ablation differentially induced senescence of BMSCs and HSCs in relation to age without alteration in their BM frequency. Our findings support that deletion of Wls in Col2.3-expressing cells induces senescence of BMSCs and impairs BM microenvironment in age-independent manner. Overall, long-term deterioration in BM microenvironment contributes to age-related HSC senescence with impaired progeny and hematopoiesis, which also suggests possible roles of osteoblastic Wls on the maintenance of BM HSCs.


Assuntos
Envelhecimento/metabolismo , Células da Medula Óssea/metabolismo , Deleção de Genes , Osteoblastos/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Células-Tronco/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/metabolismo
8.
Korean J Orthod ; 50(3): 206-215, 2020 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-32475848

RESUMO

Osteochondroma is a common benign tumor of bones, but it is rare in the mandibular condyle. With its outgrowth it manifests clinically as deviation of the mandible limitation of mouth opening, and facial asymmetry. After the tumor is diagnosed on the basis of clinical symptoms and radiographic examination including cone-beam computed tomography (CBCT) analysis, an appropriate surgery and treatment plan should be formulated. Herein, we present the case of a 44-year-old female patient who visited our dental hospital because her chin point had been deviating to the left side slowly but progressively over the last 3 years and she had difficulty masticating. Based on CBCT, she was diagnosed with skeletal Class III malocclusion accompanied by osteochondroma of the right mandibular condyle. Maxillary occlusal cant with the right side down was observed, but it was confirmed to be an extrusion of the molars associated with dental compensation. Therefore, after intrusion of the right molars with the use of temporary anchorage devices, sagittal split ramus osteotomy was used to remove the tumor and perform orthognathic surgery simultaneously. During 6 months after the surgery, continuous bone resorption and remodeling were observed in the condyle of the affected side, which led to a change in occlusion. During the postoperative orthodontic treatment, intrusive force and buccal torque were applied to the molars on the affected side, and a proper buccal overjet was created. After 18 months, CBCT revealed that the rate of bone absorption was continuously reduced, bone corticalization appeared, and good occlusion and a satisfying facial profile were achieved.

9.
Korean J Orthod ; 50(2): 136-144, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32257939

RESUMO

Hemifacial microsomia (HFM) patients may experience emotional withdrawal during their growth period due to their abnormal facial appearance. Distraction osteogenesis at an early age to improve their appearance can encourage these patients. Some abnormalities of the affected side can be overcome by distraction osteogenesis at an early age. However, differences in the growth rate between the affected and unaffected sides during the rest of the growth period are inevitable due to the characteristics of HFM. Therefore, re-evaluation should be performed after completion of growth in order to achieve stable occlusion through either orthognathic surgery or camouflage orthodontic treatment. An eight-year-old patient visited the clinic exhibiting features of HFM with slight mandibular involvement. He received phase I treatment with distraction osteogenesis and a functional appliance. Distraction osteogenesis was performed at the right ramus, which resulted in an open bite at the right posterior dentition. After distraction osteogenesis, a functional appliance and partial fixed appliance were used to achieve extrusion of the affected posterior dentition and settlement of the occlusion adjustment on the unaffected posterior dentition. The patient visited the clinic regularly for follow-up assessments, and at the age of 20 years, he showed facial asymmetry of the mandible, which had deviated to the right side. He received orthodontic treatment to improve the occlusion of his posterior dentition after the growth period. Without orthognathic surgery, stable occlusion and a satisfactory facial appearance were obtained through camouflage orthodontic treatment.

10.
J Periodontal Res ; 54(6): 690-701, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31328274

RESUMO

OBJECTIVE: Dietary bioactive materials having anti-inflammatory and antioxidant potentials are able to inhibit diabetes-associated periodontal complications. Although numerous studies indicate that administration of p-coumaric acid (p-CA) ameliorates diabetes and diabetes-related complications, the roles of p-CA on periodontal tissue destruction in diabetic mice and the possible mechanisms therein are not completely understood. In this study, we evaluated whether supplementation with p-CA protects mice against diabetes-associated spontaneous periodontal destruction and also explored the associated mechanism therein using in vivo and in vitro experimental systems. MATERIALS AND METHODS: C57BL/6 male mice were divided into sham, streptozotocin (STZ), and STZ+CA groups (n = 5/group). Sham group was intraperitoneally injected with sodium buffer, whereas other two groups were injected with the buffer containing 160 mg/kg of STZ. STZ-induced diabetic mice received oral gavage with p-CA (50 mg/kg) (STZ+CA group) or with buffer only (STZ group) daily for 6 weeks. The effect of p-CA on diabetes-associated spontaneous periodontal destruction was evaluated using µCT analysis, hematoxylin and eosin staining, tartrate-resistant acid phosphatase staining, and immunohistochemical staining methods. The efficacies of p-CA on cell proliferation, osteoblast differentiation, reactive oxygen species (ROS) accumulation, and antioxidant-related marker expression were examined using human periodontal ligament fibroblasts (hPLFs) cultured under high glucose condition. RESULTS: Streptozotocin group exhibited periodontal tissue destruction along with increased inflammation, oxidative stress, and osteoclast formation, as well as with decreased osteogenesis. However, oral administration with p-CA protected mice against STZ-induced periodontal destruction by inhibiting inflammation and osteoclastic activation. STZ+CA group also showed higher expression of antioxidant and osteogenic markers in periodontal tissue than did STZ group. Treatment with high glucose concentration (30 mmol/L) impaired proliferation and osteoblast differentiation of hPLFs along with cellular ROS accumulation, whereas these impairments were almost completely disappeared by supplementation with p-CA. CONCLUSION: These findings demonstrate that supplementation with p-CA inhibits diabetes-associated spontaneous destruction of periodontal tissue by enhancing anti-inflammatory, anti-osteoclastogenic, and antioxidant defense systems in STZ-treated mice.


Assuntos
Diabetes Mellitus Experimental/complicações , Suplementos Nutricionais , Estresse Oxidativo , Doenças Periodontais/tratamento farmacológico , Propionatos/farmacologia , Administração Oral , Animais , Antioxidantes/metabolismo , Células Cultivadas , Ácidos Cumáricos , Fibroblastos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Periodontais/etiologia , Ligamento Periodontal/citologia , Estreptozocina
11.
J Bone Miner Metab ; 37(5): 900-912, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30843129

RESUMO

Numerous studies have demonstrated the advantages of plant cell suspension culture systems in producing bioactive recombinant human growth factors. This study investigated the biological activity of recombinant basic human fibroblast growth factor (rhFGF2) protein produced by a plant culture system to enhance new bone formation in a bone defect mouse model. The human FGF2 cDNA gene was cloned into a plant expression vector driven by the rice α-amylase 3D promoter. The vector was introduced into rice calli (Oryza sativa L. cv. Dongjin), and the clone with the highest expression of rhFGF2 was selected. Maximum accumulation of rhFGF2 protein (approximately 28 mg/l) was reached at 13 day post-incubation. Male C57BL/6 mice underwent calvarial defect surgery and the defects were loaded with absorbable collagen sponge (ACS) only (ACS group) or ACS impregnated with 5 µg of plant-derived rhFGF2 (p-FGF2) protein or E. coli-derived rhFGF2 (e-FGF2) protein. Similar to the effects of e-FGF2, local delivery with p-FGF2 enhanced bone healing in the damaged region to higher levels than the ACS group. Exogenous addition of p-FGF2 or e-FGF2 exhibited similar effects on proliferation, mineralization, and osteogenic marker expression in MC3T3-E1 cells. Together, the current findings support the usefulness of this plant-based expression system for the production of biologically active rhFGF2.


Assuntos
Suplementos Nutricionais , Fator 2 de Crescimento de Fibroblastos/farmacologia , Oryza/genética , Osteogênese/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Crânio/patologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Biomarcadores/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Escherichia coli/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/isolamento & purificação , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Plantas Geneticamente Modificadas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Crânio/efeitos dos fármacos
12.
Neuromuscul Disord ; 27(6): 550-556, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28433475

RESUMO

We performed targeted population screening of late onset Pompe disease (LOPD) in unspecified myopathy patients, because early diagnosis is difficult due to its heterogeneous clinical features. We prospectively enrolled 90 unrelated myopathic patients who had one or more signs out of five LOPD-like clinical findings (proximal weakness, axial weakness, lingual weakness, respiratory difficulty, idiopathic hyperCKemia). Acid alpha glucosidase activity was evaluated with dried blood spot and mixed leukocyte simultaneously. For a final diagnosis of LOPD, 16 patients with decreased enzyme activity were genotyped by GAA molecular analysis. We found two patients with LOPD (2.2%), and the remaining 14 patients had at least one G576S or E689K mutation, known as the pseudodeficiency allele. Acid alpha glucosidase activity of LOPD patients was significantly lower than that of patients with at least one pseudodeficiency allele (p = 0.017). This study is the first LOPD screening study for targeted Korean population, and more generally, an Asian population. Our findings suggest that for diagnosis of LOPD in Asian population, modified cutoff value of acid alpha glucosidase activity with dry blood spot considering that of patients having heterozygote pathogenic variants or pseudodeficiency alleles may reduce time and cost requirements and increase the comfort of patients.


Assuntos
Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doenças Musculares/diagnóstico , Adulto , Feminino , Doença de Depósito de Glicogênio Tipo II/sangue , Doença de Depósito de Glicogênio Tipo II/complicações , Doença de Depósito de Glicogênio Tipo II/enzimologia , Humanos , Masculino , Doenças Musculares/sangue , Doenças Musculares/complicações , Doenças Musculares/enzimologia , Estudos Prospectivos , República da Coreia/epidemiologia , alfa-Glucosidases/sangue
13.
J Bone Miner Metab ; 35(5): 485-496, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27766421

RESUMO

Fibroblast growth factor 7 (FGF7) plays an important role in regulating the proliferation, migration, and differentiation of cells. However, the role of FGF7 in bone formation is not yet fully understood. We examined the effect of FGF7 on bone formation using a rat model of mandible defects. Rats underwent mandible defect surgery and then either scaffold treatment alone (control group) or FGF7-impregnated scaffold treatment (FGF7 group). Micro-CT and histological analyses revealed that the FGF7 group exhibited greater bone formation than did the control group 10 weeks after surgery. With the exception of total porosity (%), all bone parameters had higher values in the FGF7 group than in the control group at each follow-up after surgery. The FGF7 group showed greater expression of osteogenic markers, such as runt-related transcription factor 2, osterix, osteocalcin, bone morphogenetic protein 2, osteopontin, and type I collagen in newly formed bone than did the control group. The delivery of FGF7 also increased the messenger RNA expression of stromal-cell-derived factor 1 (SDF-1) and CXCR4 in newly formed bone in the FGF7 group compared with the control group. Further, addition of exogenous FGF7 induced migration of rat bone marrow stromal cells and increased the expression of SDF-1 and CXCR4 in the cells. Furthermore, the addition of FGF7 augmented mineralization in the cells with increased expression of osteogenic markers, and this augmentation was significantly suppressed by an inhibitor specific for c-Jun N-terminal kinase (SP600125) or extracellular-signal-regulated kinase (PD98059). Collectively, these results suggest that local delivery of FGF7 increases bone formation in a mandible defect with enhanced osteogenesis and chemoattraction.

14.
Korean J Orthod ; 44(5): 246-53, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25309864

RESUMO

OBJECTIVE: To evaluate the bioactivity, and the biomechanical and bone-regenerative properties of Ti6Al4V miniscrews subjected to anodization, cyclic precalcification, and heat treatment (APH treatment) and their potential clinical use. METHODS: The surfaces of Ti6Al4V alloys were modified by APH treatment. Bioactivity was assessed after immersion in simulated body fluid for 3 days. The hydrophilicity and the roughness of APH-treated surfaces were compared with those of untreated (UT) and anodized and heat-treated (AH) samples. For in vivo tests, 32 miniscrews (16 UT and 16 APH) were inserted into 16 Wistar rats, one UT and one APH-treated miniscrew in either tibia. The miniscrews were extracted after 3 and 6 weeks and their osseointegration (n = 8 for each time point and group) was investigated by surface and histological analyses and removal torque measurements. RESULTS: APH treatment formed a dense surface array of nanotubular TiO2 layer covered with a compact apatite-like film. APH-treated samples showed better bioactivity and biocompatibility compared with UT and AH samples. In vivo, APH-treated miniscrews showed higher removal torque and bone-to-implant contact than did UT miniscrews, after both 3 and 6 weeks (p < 0.05). Also, early deposition of densely mineralized bone around APH-treated miniscrews was observed, implying good bonding to the treated surface. CONCLUSIONS: APH treatment enhanced the bioactivity, and the biomechanical and bone regenerative properties of the Ti6Al4V alloy miniscrews. The enhanced initial stability afforded should be valuable in orthodontic applications.

15.
J Periodontol ; 85(4): 645-54, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23805819

RESUMO

BACKGROUND: Periodontal ligament fibroblasts (PLFs) maintain homeostasis of periodontal ligaments by producing paracrine factors that affect various functions of stem-like cells. It is hypothesized that PLFs induce proliferation and differentiation of stem cells more effectively than gingival fibroblasts (GFs) and skin fibroblasts (SFs). METHODS: PLFs and GFs were isolated from extracted teeth and cultured in the presence and absence of osteogenesis-inducing factors. Mouse embryonic stem (mES) cells and SFs were purchased commercially. mES cells were incubated with culture supernatants of these fibroblasts or cocultured directly with the cells. Proliferation and mineralization in mES cells were determined at various times of incubation. Immunostaining and polymerase chain reaction were performed. The activity of mitogen-activated protein kinase and alkaline phosphatase (ALP) was also measured. RESULTS: In cocultures, PLFs stimulated proliferation of mES cells more effectively than GFs or SFs. Similarly, the addition of culture supernatant of PLFs induced the most prominent proliferation of mES cells, and this was significantly inhibited by treatment with antibody against fibroblast growth factor (FGF)4 or the c-Jun N-terminal kinase inhibitor SP600125 (anthra[1,9-cd]pyrazol-6(2H)-one). Supplementation with culture supernatant from the fibroblasts induced osteogenic differentiation of mES cells in the order PLFs > GFs > SFs. These activities of PLFs were related to their potential to produce osteogenic markers, such as ALP and runt-related transcription factor-2 (Runx2), and to secrete FGF7. Pretreatment of mES cells with the extracellular signal-regulated kinase inhibitor PD98059 [2-(2-amino-3-methyoxyphenyl)-4H-1-benzopyran-4-one] or SP600125 clearly attenuated mineralization induced by culture supernatant of PLF with attendant decreases in mRNA levels of Runx2, bone sialoprotein, osteocalcin, and osteopontin. CONCLUSION: PLFs regulate the proliferation and osteogenic differentiation of mES cells more strongly than GFs and SFs via the secretion of FGF through a mechanism that involves mitogen-activated protein kinase-mediated signaling.


Assuntos
Células-Tronco Embrionárias/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Fibroblastos/fisiologia , Osteogênese/fisiologia , Ligamento Periodontal/citologia , Fosfatase Alcalina/análise , Animais , Antracenos/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Técnicas de Cocultura , Subunidade alfa 1 de Fator de Ligação ao Core/análise , Meios de Cultivo Condicionados , Fator 4 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 7 de Crescimento de Fibroblastos/análise , Fatores de Crescimento de Fibroblastos/análise , Flavonoides/farmacologia , Gengiva/citologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/análise , Osteocalcina/análise , Osteopontina/análise , Pele/citologia
16.
Mol Cell Biochem ; 382(1-2): 37-45, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24026476

RESUMO

Fibroblast growth factor-7 (FGF7) is known to regulate proliferation and differentiation of cells; however, little information is available on how FGF7 affects the differentiation of embryonic stem cells (ESCs). We examined the effects of FGF7 on proliferation and osteogenic differentiation of mouse ESCs. Exogenous FGF7 addition did not change the proliferation rate of mouse ESCs. In contrast, the addition of FGF7 facilitated the dexamethasone, ascorbic acid, and ß-glycerophosphate (DAG)-induced increases in bone-like nodule formation and calcium accumulation. FGF7 also augmented mRNA expression of runt-related transcription factor-2 (Runx2), osterix, bone sialoprotein (BSP), and osteocalcin (OC) in the presence of DAG. FGF7-mediated increases in the mineralization and bone-specific gene expression were almost completely attenuated by pretreating with anti-FGF7 antibody. FGF7 treatment accelerated the DAG-induced activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in the cells. A pharmacological inhibitor specific to ERK, but not to JNK or p38 kinase, dramatically suppressed FGF7-mediated mineralization and accumulation of collagen and OC in the presence of DAG. This suppression was accompanied by the reduction in Runx2, osterix, BSP, and OC mRNA levels, which were increased by FGF7 in the presence of DAG. Collectively, our results suggest that FGF7 stimulates osteogenic differentiation, but not proliferation, in ESCs, by activating ERK/Runx2 signaling.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 7 de Crescimento de Fibroblastos/farmacologia , Osteogênese/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/genética , Diferenciação Celular/genética , Linhagem Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glicerofosfatos/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Osteogênese/genética , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
PLoS One ; 8(8): e71641, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967228

RESUMO

Fibroblast growth factor-4 (FGF4) is expressed in embryonic stages and in adult tissues, where it plays critical roles in modulating multiple cellular functions. However, the exact roles of FGF4 on proliferation and differentiation of embryonic stem cells (ESCs) are not completely understood. Exogenous addition of FGF4 stimulated proliferation of mouse ESCs (mESCs), as proven by the increases in DNA synthesis and cell cycle regulatory protein induction. These increases were almost completely inhibited by pre-treating cells with anti-FGF4 antibody. FGF4 also activated c-Jun N-terminal kinase (JNK) and extracellular-signal regulated kinase (ERK) signaling, but not p38 kinase. Blockage of JNK signaling by SP600125 or by transfection with its specific siRNA significantly inhibited FGF4-stimulated cell proliferation through the suppression of c-Jun induction and activator protein-1 (AP-1) activity. However, ERK or p38 kinase inhibitor did not affect FGF4-stimulated proliferation in mESCs. FGF4 suppressed osteogenic differentiation of mESCs by inhibiting expression of transcription factors involved in bone formation. Further, exogenous FGF4 addition stimulated proliferation of human periodontal ligament stem cells (hPDLSCs) and bone marrow mesenchymal stem cells (BMMSCs) via activation of ERK signaling. FGF4 also augmented mineralization of hPDLSCs, but not of BMMSCs. Collectively, it is suggested that FGF4 triggers proliferation of stem cells by activating MAPK-mediated signaling, while it affects differently osteogenic differentiation according to the origins of stem cells.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Fator 4 de Crescimento de Fibroblastos/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Calcificação Fisiológica/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células-Tronco Embrionárias/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 4 de Crescimento de Fibroblastos/antagonistas & inibidores , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fosforilação , Ligação Proteica , Fator de Transcrição Sp7 , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
BMB Rep ; 45(10): 571-6, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23101511

RESUMO

Radiotherapy is considered to cause detrimental effects on bone tissue eventually increasing bone loss and fracture risk. However, there is a great controversy on the real effects of irradiation itself on osteoblasts, and the mechanisms by which irradiation affects osteoblast differentiation and mineralization are not completely understood. We explored how X-ray radiation influences differentiation and bone-specific gene expression in mouse calvarial osteoblasts. Irradiation at 2 Gy not only increased differentiation and mineralization of the cells, but also upregulated the expression of alkaline phosphatase, type I collagen, osteopontin, and osteocalcin at early stages of differentiation. However, irradiation at higher doses (>2 Gy) did not stimulate osteoblast differentiation, rather it suppressed DNA synthesis by the cells without a toxic effect. Additional experiments suggested that transforming growth factor-beta 1 and runt-transcription factor 2 play important roles in irradiation- stimulated bone differentiation by acting as upstream regulators of bone-specific markers.


Assuntos
Diferenciação Celular/efeitos da radiação , Osteoblastos/efeitos da radiação , Radiação Ionizante , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Calcificação Fisiológica/efeitos da radiação , Células Cultivadas , 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 , Citocinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , RNA Mensageiro/metabolismo , Crânio/citologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima
19.
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
20.
J Cell Biochem ; 113(5): 1724-32, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22213029

RESUMO

The antigen I/II (AgI/II) protein is a major surface protein that mediates the attachment of Streptococcus mutans (S. mutans) to the saliva-coated pellicle. Numerous studies have investigated not only the mechanisms by which AgI/II signaling is transduced within cells, but have also attempted to use AgI/II-specific antibodies to treat dental caries and host immune responses. However, little information is available about the effects of AgI/II on basic cellular events in bone cells. In this study, we examined the effects of the His-tagged recombinant N-terminal half of the AgI/II protein (rAgI/II-N) generated from S. mutans GS-5 on the viability, proliferation, and cell cycle progression of primary calvarial osteoblasts. We also investigated the mechanisms involved in the rAgI/II-N-mediated survival of serum-starved osteoblasts. We found that rAgI/II treatment attenuated the serum deprivation-induced decrease in cell viability and proliferation of osteoblasts. rAgI/II-N also prevented the loss of mitochondrial membrane potential (MMP), alterations in levels of two key mitochondrial Bcl-2 family proteins, and the accumulation of numerous cells into the sub-G(1) phase that were observed in serum-starved osteoblasts. Pharmacological inhibitors of phosphoinositide 3-kinase (PI3K), but not of extracellular signal-regulated kinase or Ras, blocked the rAgI/II-N-mediated protection against serum deprivation-induced cell death. Additional experiments revealed that the integrin α5ß1-mediated PI3K pathway is required for rAgI/II-N-mediated Akt phosphorylation in osteoblasts. Collectively, these results suggest that rAgI/II-N induces survival signals in serum-starved osteoblasts through integrin-induced PI3K/Akt signaling pathways.


Assuntos
Antígenos de Bactérias/fisiologia , Sobrevivência Celular/imunologia , Osteoblastos/microbiologia , Osteoblastos/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Streptococcus mutans/imunologia , Animais , Antígenos de Bactérias/administração & dosagem , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/imunologia , Ciclo Celular , Proliferação de Células , Células Cultivadas , Meios de Cultura Livres de Soro , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Mitocôndrias/metabolismo , Modelos Biológicos , Osteoblastos/imunologia , Osteoblastos/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Transdução de Sinais , Streptococcus mutans/patogenicidade , Estresse Fisiológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA