Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Clin Oral Investig ; 27(5): 2335-2346, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36607492

RESUMEN

OBJECTIVE: This study was aimed to delineate the clinical, CBCT radiographic characteristics, and complications of maxillary molar in a periodontitis population. MATERIALS AND METHODS: Medical records and CBCT images were utilized to identify adult patients with periodontitis in a tertiary referral dental hospital between June 2019 and December 2020. CBCT scan images were used to characterize the detailed bone thickness, absorbing height, and position of maxillary molar as well as their associated conditions. All relevant descriptive epidemiological data, clinical information, radiographic details, and associated complications were recorded and statistically analyzed. RESULTS: According to the above criteria, 577 eligible periodontitis patients were enrolled and defined as research cohort here with mean age 45 ± 4.8 years. Male patients outnumbered females with a gender ratio of 1.23:1. Our results demonstrated that the bone loss of maxillary first molar was more serious than that of second molar with tooth position symmetry. The occurrence of various complications (periodontal abscess, pulp lesions, furcation lesion, and mucosal thickening) was significantly correlated to periodontal-related clinical parameters of maxillary molar. CONCLUSIONS: Our results demonstrated the more serious bone loss of maxillary first molar with tooth position symmetry. The occurrence of various complications was significantly correlated to periodontal-related clinical parameters. Our findings offer valuable information concerning the clinical, radiographic characteristics, and complications of maxillary molar in a periodontitis population. CLINICAL RELEVANCE: These findings are beneficial for clinicians to comprehensively understand the bone status, pathogenesis, and clinical management of maxillary molar in periodontitis.


Asunto(s)
Periodontitis , Tomografía Computarizada de Haz Cónico Espiral , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Tomografía Computarizada de Haz Cónico/métodos , Periodontitis/diagnóstico por imagen , Periodontitis/patología , Diente Molar/diagnóstico por imagen , Diente Molar/patología
2.
Exp Cell Res ; 372(2): 158-167, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30268758

RESUMEN

Trio, the Rho guanine nucleotide exchange factor (Rho-GEF), plays diverse roles in cell migration, cell axon guidance and cytoskeleton reorganization. Conserved during evolution, Trio encodes two guanine nucleotide exchange factor domains (GEFs) and activates small GTPases. The Rho-family small GTPases RhoA and Rac1, which are target molecules of Trio, have been described to engage in craniofacial development and tooth formation. However, the exact role of Trio in tooth development remains elusive. In this study, we generated Wnt1-cre;Triofl/fl mice to address the potential function of Trio in tooth development. Wnt1-cre;Triofl/fl mice showed short root deformity as well as decreased expression of odontogenic makers such as RUNX2, OSX, OCN, and OPN. In vitro, Trio was silenced in human stem cells of dental papilla (SCAPs). Compared with the control group, the proliferation and migration ability in the experimental group was disrupted. After knocking down Trio in SCAPs, the cells showed phenotypes of poor odontogenic differentiation and weak mineralized nodules. To study the underlying mechanism, we investigated the p38 MAPK pathway and found that loss of Trio blocked the cascade transduction of p38 MAPK signaling. In conclusion, we identified Trio as a novel coordinator in regulating root development and clarified its relevant molecular events.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Odontogénesis/genética , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Raíz del Diente/crecimiento & desarrollo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Papila Dental/crecimiento & desarrollo , Papila Dental/metabolismo , Humanos , Ratones , Neuropéptidos/genética , Unión Proteica/genética , Transducción de Señal/genética , Células Madre/citología , Células Madre/metabolismo , Raíz del Diente/metabolismo , Proteína de Unión al GTP rac1/genética , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA
3.
Int J Oral Sci ; 15(1): 26, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37380627

RESUMEN

Periodontitis imparting the increased risk of atherosclerotic cardiovascular diseases is partially due to the immune subversion of the oral pathogen, particularly the Porphyromonas gingivalis (P. gingivalis), by inducing apoptosis. However, it remains obscure whether accumulated apoptotic cells in P. gingivalis-accelerated plaque formation are associated with impaired macrophage clearance. Here, we show that smooth muscle cells (SMCs) have a greater susceptibility to P. gingivalis-induced apoptosis than endothelial cells through TLR2 pathway activation. Meanwhile, large amounts of miR-143/145 in P.gingivalis-infected SMCs are extracellularly released and captured by macrophages. Then, these miR-143/145 are translocated into the nucleus to promote Siglec-G transcription, which represses macrophage efferocytosis. By constructing three genetic mouse models, we further confirm the in vivo roles of TLR2 and miR-143/145 in P. gingivalis-accelerated atherosclerosis. Therapeutically, we develop P.gingivalis-pretreated macrophage membranes to coat metronidazole and anti-Siglec-G antibodies for treating atherosclerosis and periodontitis simultaneously. Our findings extend the knowledge of the mechanism and therapeutic strategy in oral pathogen-associated systemic diseases.


Asunto(s)
Aterosclerosis , MicroARNs , Animales , Ratones , Células Endoteliales , Receptor Toll-Like 2 , Macrófagos , Apoptosis , Miocitos del Músculo Liso
4.
Theranostics ; 11(9): 4316-4334, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754063

RESUMEN

Trio is a unique member of the Rho-GEF family that has three catalytic domains and is vital for various cellular processes in both physiological and developmental settings. TRIO mutations in humans are involved in craniofacial abnormalities, in which patients present with mandibular retrusion. However, little is known about the molecular mechanisms of Trio in neural crest cell (NCC)-derived craniofacial development, and there is still a lack of direct evidence to assign a functional role to Trio in NCC-induced craniofacial abnormalities. Methods:In vivo, we used zebrafish and NCC-specific knockout mouse models to investigate the phenotype and dynamics of NCC development in Trio morphants. In vitro, iTRAQ, GST pull-down assays, and proximity ligation assay (PLA) were used to explore the role of Trio and its potential downstream mediators in NCC migration and differentiation. Results: In zebrafish and mouse models, disruption of Trio elicited a migration deficit and impaired the differentiation of NCC derivatives, leading to craniofacial growth deficiency and mandibular retrusion. Moreover, Trio positively regulated Myh9 expression and directly interacted with Myh9 to coregulate downstream cellular signaling in NCCs. We further demonstrated that disruption of Trio or Myh9 inhibited Rac1 and Cdc42 activity, specifically affecting the nuclear export of ß-catenin and NCC polarization. Remarkably, craniofacial abnormalities caused by trio deficiency in zebrafish could be partially rescued by the injection of mRNA encoding myh9, ca-Rac1, or ca-Cdc42. Conclusions: Here, we identified that Trio, interacting mostly with Myh9, acts as a key regulator of NCC migration and differentiation during craniofacial development. Our results indicate that trio morphant zebrafish and Wnt1-cre;Triofl/fl mice offer potential model systems to facilitate the study of the pathogenic mechanisms of Trio mutations causing craniofacial abnormalities.


Asunto(s)
Cadenas Pesadas de Miosina/genética , Cresta Neural/fisiología , Animales , Diferenciación Celular/genética , Línea Celular , Movimiento Celular/genética , Embrión de Mamíferos/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , Transducción de Señal/genética , Pez Cebra , beta Catenina/genética
5.
Int J Biol Sci ; 17(15): 4238-4253, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34803495

RESUMEN

Background: Congenital anomalies are increasingly becoming a global pediatric health concern, which requires immediate attention to its early diagnosis, preventive strategies, and efficient treatments. Guanine nucleotide binding protein, alpha inhibiting activity polypeptide 3 (Gnai3) gene mutation has been demonstrated to cause congenital small jaw deformity, but the functions of Gnai3 in the disease-specific microRNA (miRNA) upregulations and their downstream signaling pathways during osteogenesis have not yet been reported. Our previous studies found that the expression of Mir24-2-5p was significantly downregulated in the serum of young people with overgrowing mandibular, and bioinformatics analysis suggested possible binding sites of Mir24-2-5p in the Gnai3 3'UTR region. Therefore, this study was designed to investigate the mechanism of Mir24-2-5p-mediated regulation of Gnai3 gene expression and explore the possibility of potential treatment strategies for bone defects. Methods: Synthetic miRNA mimics and inhibitors were transduced into osteoblast precursor cells to regulate Mir24-2-5p expression. Dual-luciferase reporter assay was utilized to identify the direct binding of Gnai3 and its regulator Mir24-2-5p. Gnai3 levels in osteoblast precursor cells were downregulated by shRNA (shGnai3). Agomir, Morpholino Oligo (MO), and mRNA were microinjected into zebrafish embryos to control mir24-2-5p and gnai3 expression. Relevant expression levels were determined by the qRT-PCR and Western blotting. CCK-8 assay, flow cytometry, and transwell migration assays were performed to assess cell proliferation, apoptosis, and migration. ALP, ARS and Von Kossa staining were performed to observe osteogenic differentiation. Alcian blue staining and calcein immersions were performed to evaluate the embryonic development and calcification of zebrafish. Results: The expression of Mir24-2-5p was reduced throughout the mineralization process of osteoblast precursor cells. miRNA inhibitors and mimics were transfected into osteoblast precursor cells. Cell proliferation, migration, osteogenic differentiation, and mineralization processes were measured, which showed a reverse correlation with the expression of Mir24-2-5p. Dual-luciferase reporter gene detection assay confirmed the direct interaction between Mir24-2-5p and Gnai3 mRNA. Moreover, in osteoblast precursor cells treated with Mir24-2-5p inhibitor, the expression of Gnai3 gene was increased, suggesting that Mir24-2-5p negatively targeted Gnai3. Silencing of Gnai3 inhibited osteoblast precursor cells proliferation, migration, osteogenic differentiation, and mineralization. Promoting effects of osteoblast precursor cells proliferation, migration, osteogenic differentiation, and mineralization by low expression of Mir24-2-5p was partially rescued upon silencing of Gnai3. In vivo, mir24-2-5p Agomir microinjection into zebrafish embryo resulted in shorter body length, smaller and retruded mandible, decreased cartilage development, and vertebral calcification, which was partially rescued by microinjecting gnai3 mRNA. Notably, quite similar phenotypic outcomes were observed in gnai3 MO embryos, which were also partially rescued by mir24-2-5p MO. Besides, the expression of phospho-JNK (p-JNK) and p-p38 were increased upon Mir24-2-5p inhibitor treatment and decreased upon shGnai3-mediated Gnai3 downregulation in osteoblast precursor cells. Osteogenic differentiation and mineralization abilities of shGnai3-treated osteoblast precursor cells were promoted by p-JNK and p-p38 pathway activators, suggesting that Gnai3 might regulate the differentiation and mineralization processes in osteoblast precursor cells through the MAPK signaling pathway. Conclusions: In this study, we investigated the regulatory mechanism of Mir24-2-5p on Gnai3 expression regulation in osteoblast precursor cells and provided a new idea of improving the prevention and treatment strategies for congenital mandibular defects and mandibular protrusion.


Asunto(s)
Diferenciación Celular/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , MicroARNs/metabolismo , Osteoblastos/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Regulación de la Expresión Génica/fisiología , MAP Quinasa Quinasa 4/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Imitación Molecular , ARN/química , ARN/farmacología , Transducción de Señal , Regulación hacia Arriba , Pez Cebra , Proteínas Quinasas p38 Activadas por Mitógenos/genética
6.
Int J Biol Sci ; 16(1): 181-193, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31892855

RESUMEN

Tooth development is a complex process that is regulated precisely by several signalling pathways and transcription factors. GATA-binding protein 4 (GATA4) is a DNA binding transcription factor, and our previous study showed that GATA4 is a novel regulator of root development. However, it remains unclear whether GATA4 is necessary for odontoblast differentiation and dentin formation. Here, we evaluated the phenotypic changes of Wnt1-Cre;GATA4fl/fl mice. The mutant mice showed defective dentin and short root deformity. The odontoblasts lost polarity instead of exhibiting a shorter height and flattened morphology. Moreover, the expression of several molecules, such as DSPP, COL-1, DCN, and PCNA, were downregulated during mutant tooth development. In vivo, we injected lentivirus to overexpress GATA4 in mice root. The dentin formation and the expression of odonto/osteogenic markers (DSPP, COL-1, DCN) were enhanced in the GATA4 overexpression group. During the in vitro study, the ability of proliferation, migration and odonto/osteogenic differentiation was declined by GATA4 knockdown approach in human dental pulp stem cells (DPSCs). The expression of odonto/osteogenic markers (DSPP, BMP4, RUNX2, OSX, OPN, OCN) was reduced in the shGATA4 group, while overexpressing GATA4 in DPSCs promoted mineralization. Furthermore, an immunoprecipitation-mass spectrometry procedure was used to confirm the interaction between GATA4 and Fructose-1, 6-bisphosphatase 1 (FBP1). We used gain and lose-of-function to delineated the role of GATA4 in regulating FBP1 expression. Knocking down GATA4 in DPSCs resulted in decreased glucose consumption and lactate production. We used small hairpin RNA targeting FBP1 to reduce the expression of FBP1 in DPSCs, which significantly increased glucose consumption and lactate production. Together, the results suggested that GATA4 is important for root formation and odontoblast polarity, as it promotes the growth and differentiation of dental mesenchymal cells around the root and affects the glucose metabolism of DPSCs via the negative regulation of FBP1.


Asunto(s)
Dentina/metabolismo , Fructosa-Bifosfatasa/metabolismo , Factor de Transcripción GATA4/metabolismo , Raíz del Diente/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Dentinogénesis/genética , Dentinogénesis/fisiología , Fructosa-Bifosfatasa/genética , Factor de Transcripción GATA4/genética , Gluconeogénesis/genética , Gluconeogénesis/fisiología , Ratones Noqueados , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Odontoblastos/citología , Odontoblastos/metabolismo
7.
Int J Biol Sci ; 15(12): 2538-2547, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31754327

RESUMEN

Background/Aims: Neural crest cells play a vital role in craniofacial development, microRNA-1 (miR-1) is essential in development and disease of the cardiac and skeletal muscle, the objective of our study is to investigate effects of miR-1 on neural crest cell in the craniofacial development and its molecular mechanism. Methods: We knocked down miR-1 in zebrafish by miR-1 morpholino (MO) microinjection and observed phenotype of neural crest derivatives. We detected neural crest cell migration by time-lapse. Whole-mount in situ hybridization was used to monitor the expressions of genes involved in neural crest cell induction, specification, migration and differentiation. We performed a quantitative proteomics study (iTRAQ) and bioinformatics prediction to identify the targets of miR-1 and validate the relationship between miR-1 and its target gene sec63. Results: We found defects in the tissues derived from neural crest cells: a severely reduced lower jaw and delayed appearance of pigment cells. miR-1 MO injection also disrupted neural crest cell migration. At 24 hours post fertilization (hpf), reduced expression of tfap2a, dlx2, dlx3b, ngn1 and crestin indicated that miR-1 deficiency affected neural crest cell differentiation. iTRAQ and luciferase reporter assay identified SEC63 as a direct target gene of miR-1. The defects of miR-1 deficiency could be reversed, at least in part, by specific suppression of sec63 expression. Conclusion: miR-1 is involved in the regulation of neural crest cell development, and that it acts, at least partially, by targeting sec63 expression.


Asunto(s)
Desarrollo Maxilofacial/genética , Proteínas de la Membrana/genética , MicroARNs/fisiología , Cresta Neural/crecimiento & desarrollo , Proteínas de Unión al ARN/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Diferenciación Celular , Movimiento Celular , Biología Computacional , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hibridación in Situ , MicroARNs/genética , MicroARNs/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Proteómica , Cráneo/embriología , Imagen de Lapso de Tiempo , Pez Cebra/genética , Pez Cebra/metabolismo
8.
Int J Mol Med ; 43(1): 382-392, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30431055

RESUMEN

Odonto/osteogenic differentiation of stem cells from the apical papilla (SCAPs) is a key process in tooth root formation and development. However, the molecular mechanisms underlying this process remain largely unknown. In the present study, it was identified that guanine and nucleotide binding protein 3 (GNAI3) was at least in part responsible for the odonto/osteogenic differentiation of SCAPs. GNAI3 was markedly induced in mouse tooth root development in vivo and in human SCAPs mineralization in vitro. Notably, knockdown of GNAI3 by lentiviral vectors expressing short­hairpin RNAs against GNAI3 significantly inhibited the proliferation, cell cycle progression and migration of SCAPs, as well as odonto/osteogenic differentiation of SCAPs in vitro, suggesting that GNAI3 may play an essential role in tooth root development. The promotive role of GNAI3 in odonto/osteogenic differentiation was further confirmed by downregulation of odonto/osteogenic makers in GNAI3­deficient SCAPs. In addition, knockdown of GNAI3 effectively suppressed activity of c­Jun N­terminal kinase (JNK) and extracellular­signal regulated kinase (ERK) signaling pathways that was induced during SCAPs differentiation, suggesting that GNAI3 promotes SCAPs mineralization at least partially via JNK/ERK signaling. Taken together, the present results implicate GNAI3 as a critical regulator of odonto/osteogenic differentiation of SCAPs in tooth root development, and suggest a possible role of GNAI3 in regeneration processes in dentin or other tissues.


Asunto(s)
Diferenciación Celular , Papila Dental/citología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Odontogénesis , Osteogénesis , Células Madre/enzimología , Animales , Antracenos/farmacología , Biomarcadores/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Odontogénesis/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Raíz del Diente/embriología , Raíz del Diente/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
9.
Int J Mol Med ; 41(2): 729-738, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29207140

RESUMEN

Platelet-rich plasma (PRP) is used in the clinic as an autologous blood product to stimulate bone regeneration and chondrogenesis. Numerous studies have demonstrated that PRP affects bone remodeling by accelerating osteoblast formation. With the research perspective focusing on osteoclasts, the present study established a mouse model of mandibular advancement to examine the effect of PRP on osteoclast differentiation induced by modification of the dynamics of the temporomandibular joint (TMJ). The lower incisors of the mice were trimmed by 1 mm and the resultant change in mandibular position during the process of eating induced condylar adaptation to this change. PRP significantly increased the bone mass and decreased osteoclastic activity, in vitro as well as in vivo. Mechanistically, the reduced expression of receptor activator of nuclear factor-κB ligand (RANKL)­induced differentiation marker genes, including nuclear factor of activated T-cells, cytoplasmic 1, c-fos and tartrate-resistant acid phosphatase, and that of the resorptive activity marker genes such as cathepsin k, carbonic anhydrase 2 and matrix metalloproteinase 9, indicated that PRP suppresses RANKL-induced osteoclast differentiation. A microarray analysis revealed that several genes associated with the Wnt pathway were differentially expressed, which indicated the involvement of this pathway in osteoclast differentiation. Furthermore, the activation of the Wnt pathway was verified by reverse transcription-quantitative polymerase chain reaction and immunoblot analysis of Dickkopf-related protein 1 and ß-catenin. The results of the present study indicated that PRP inhibits osteoclast differentiation through activation of the Wnt pathway.


Asunto(s)
Remodelación Ósea/genética , Diferenciación Celular/genética , Plasma Rico en Plaquetas , Ligando RANK/genética , Animales , Resorción Ósea/genética , Resorción Ósea/patología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Análisis por Micromatrices , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Linfocitos T/efectos de los fármacos , Fosfatasa Ácida Tartratorresistente/genética , Vía de Señalización Wnt/genética , beta Catenina/genética
10.
Cell Death Differ ; 25(11): 1996-2009, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29523871

RESUMEN

The role of GATA-binding protein 4 (GATA4) in neural crest cells (NCCs) is poorly defined. Here we showed that mouse NCCs lacking GATA4 exhibited developmental defects in craniofacial bone, teeth, and heart. The defects likely occurred due to decreased cell proliferation at the developmental stage. The in vitro results were consistent with the mouse model. The isobaric tags for relative and absolute quantitation assay revealed that BARX1 is one of the differentially expressed proteins after GATA4 knockdown in NCCs. On the basis of the results of dual-luciferase, electro-mobility shift, and chromatin immunoprecipitation assays, Barx1 expression is directly regulated by GATA4 in NCCs. In zebrafish, gata4 knockdown affects the development of NCCs derivatives. However, the phenotype in zebrafish could be partly rescued by co-injection of gata4 morpholino oligomers and barx1 mRNA. This study identified new downstream targets of GATA4 in NCCs and uncovered additional evidence of the complex regulatory functions of GATA4 in NCC development.


Asunto(s)
Huesos Faciales/crecimiento & desarrollo , Factor de Transcripción GATA4/metabolismo , Proteínas de Homeodominio/metabolismo , Cresta Neural/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Animales , Proliferación Celular , Huesos Faciales/diagnóstico por imagen , Huesos Faciales/metabolismo , Femenino , Factor de Transcripción GATA4/antagonistas & inhibidores , Factor de Transcripción GATA4/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Cresta Neural/citología , Cresta Neural/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción/genética , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Microtomografía por Rayos X , Pez Cebra/metabolismo
11.
J Mol Histol ; 48(3): 187-197, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28393293

RESUMEN

Osteoblasts play a major role in bone remodeling and are regulated by transcription factors. GATA4, a zinc finger transcription factor from the GATA family, has an unclear role in osteoblast differentiation. In this study, the role of GATA4 in osteoblast differentiation was studied both in vitro and in vivo by GATA4 knockdown. GATA4 expression increased during osteoblast differentiation. GATA4 knockdown in osteoblast precursor cells reduced alkaline phosphatase activity and decreased the formation of calcified nodule in an osteogenic-induced cell culture system. In vivo, micro-CT showed that local injection of lentivirus-delivered GATA4 shRNA caused reduced new bone formation during tooth movement. Histological analyses such as total collagen and Goldner's trichrome staining confirmed these results. In vivo immunohistochemical analysis showed reduced expression of osterix (OSX), osteopontin (OPN), and osteocalcin (OCN) in the shGATA4 group (P < 0.05). Consistently, both western blotting and quantitative reverse-transcription PCR proved that expression of osteogenesis-related genes, including OSX, OPN, and OCN, was significantly repressed in the shGATA4 group in vitro (P < 0.01). For further analysis of the pathways involved in this process, we examined the MAPK signaling pathway, and found knockdown of GATA4, downregulated p38 signaling pathways (P < 0.01). Collectively, these results imply GATA4 is a regulator of osteoblastic differentiation via the p38 signaling pathways.


Asunto(s)
Remodelación Ósea , Diferenciación Celular , Factor de Transcripción GATA4/fisiología , Sistema de Señalización de MAP Quinasas , Osteoblastos/citología , Animales , Células Cultivadas , Factor de Transcripción GATA4/genética , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Osteoblastos/fisiología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología
12.
J Mol Histol ; 48(5-6): 389-401, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28986711

RESUMEN

During tooth root development, stem cells from apical papillae (SCAPs) are indispensable, and their abilities of proliferation, migration and odontoblast differentiation are linked to root formation. Leucine-rich repeat-containing GPCR 4 (LGR4) modulates the biological processes of proliferation and differentiation in multiple stem cells. In this study, we showed that LGR4 is expressed in all odontoblast cell lineage cells and Hertwig's epithelial root sheath (HERS) during the mouse root formation in vivo. In vitro we determined that LGR4 is involved in the Wnt/ß-catenin signaling pathway regulating proliferation and odonto/osteogenic differentiation of SCAPs. Quantitative reverse-transcription PCR (qRT-PCR) confirmed that LGR4 is expressed during odontogenic differentiation of SCAPs. CCK8 assays and in vitro scratch tests, together with cell cycle flow cytometric analysis, demonstrated that downregulation of LGR4 inhibited SCAPs proliferation, delayed migration and arrested cell cycle progression at the S and G2/M phases. ALP staining revealed that blockade of LGR4 decreased ALP activity. QRT-PCR and Western blot analysis demonstrated that LGR4 silencing reduced the expression of odonto/osteogenic markers (RUNX2, OSX, OPN, OCN and DSPP). Further Western blot and immunofluorescence studies clarified that inhibition of LGR4 disrupted ß-catenin stabilization. Taken together, downregulation of LGR4 gene expression inhibited SCAPs proliferation, migration and odonto/osteogenic differentiation by blocking the Wnt/ß-catenin signaling pathway. These results indicate that LGR4 might play a vital role in SCAPs proliferation and odontoblastic differentiation.


Asunto(s)
Diferenciación Celular , Papila Dental/citología , Osteogénesis , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Movimiento Celular , Proliferación Celular , Forma de la Célula , Regulación hacia Abajo/genética , Silenciador del Gen , Humanos , Ratones , Morfogénesis , Odontogénesis/genética , Osteogénesis/genética , Estabilidad Proteica , ARN Interferente Pequeño/metabolismo , Células Madre/metabolismo , Raíz del Diente/citología , Raíz del Diente/crecimiento & desarrollo , Raíz del Diente/metabolismo , beta Catenina/metabolismo
13.
Sci Rep ; 7(1): 1534, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28484278

RESUMEN

Transcription factor GATA4 regulates cardiac and osteoblast differentiation. However, its role in tooth development is not clear. Therefore, we generated Wnt1-Cre;GATA4 fl/fl mice, with conditional inactivation of the GATA4 gene in the dental papilla mesenchymal cells. Phenotypic analysis showed short root deformity along with reduced expressions of odonto/osteogenic markers. Proliferation (but not apoptosis) of cells around the apical area of the root was attenuated. In vitro, we knocked down GATA4 expression in stem cells of dental apical papilla (SCAPs). Proliferation, migration and odonto/osteogenic differentiation of SCAPs were affected in the shGATA4 group. Overexpression of GATA4 in SCAPs increased mineralization. Based on our previous iTRAQ results, guanine nucleotide binding proteins 3 (GNAI3) is one of the distinct proteins after GATA4 deletion. G protein signaling is involved in bone development, remodeling, and disease. In this study, both GATA4 deletion in the mouse root and knock-down in human SCAPs decreased the expression of GNAI3. Dual-luciferase and ChIP assay confirmed the direct binding of GATA4 to the GNAI3 promoter, both in vitro and in vivo. GNAI3 knock-down significantly decreased the odonto/osteogenic differentiation ability of SCAPs. We thus establish the role of GATA4 as a novel regulator of root development and elucidate its downstream molecular events.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Odontogénesis , Adolescente , Animales , Apoptosis , Secuencia de Bases , Diferenciación Celular/genética , Movimiento Celular , Proliferación Celular , Papila Dental/patología , Femenino , Factor de Transcripción GATA4/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Humanos , Mesodermo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Cresta Neural/metabolismo , Osteogénesis/genética , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Células Madre/metabolismo , Raíz del Diente/anomalías , Raíz del Diente/patología , Regulación hacia Arriba/genética , Adulto Joven
14.
J Mol Histol ; 48(3): 199-208, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28409326

RESUMEN

Corticision is a common technique to accelerate orthodontic tooth movement; however, not much is known about the underlying mechanisms. In this study, we investigated the mechanism of alveolar tissue remodeling after corticision in a rat model of tooth movement (TM) by analyzing the differential transcriptome. A total of 36 male rats were equally divided into TM and TM with corticision (TM+C) groups. Alveolar bone response was examined using micro-computed tomography (micro-CT). Osteoclasts and osteoblasts were quantified on tartrate-resistant acid phosphatase (TRAP) and Goldner's trichrome staining. The transcriptomes of alveolus around the left maxillary first molar were determined on RNA sequencing (RNA-Seq), and the expression of selected differentially expressed genes (DEGs) validated on quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR). Immunohistochemical examination of alveolar tissue was performed to examine the expressions of correlative proteins of the selected signaling pathway in the TM and TM+C groups. The ratio of bone volume to total volume (BV/TV), and the trabecular number (Tb.N) were significantly decreased, while the movement distance and the trabecular separation (Tb.Sp) was significantly increased in the TM+C group. However, no significant between-group difference in trabecular thickness (Tb.Th) was observed. On histomorphometric analysis, a significant increase in the number of osteoclasts and increased bone resorption was observed in the TM+C group. A total of 399 DEGs were identified on RNA-SEq. Eleven selected genes were confirmed on qRT-PCR, which included components of the Ras signaling pathway. Four proteins of the Ras signaling pathway showed a higher expression in the TM+C group. Our findings indicate that corticision may speed up orthodontic tooth movement by accelerating osteoclastogenesis mediated via the Ras signaling pathway.


Asunto(s)
Remodelación Ósea/genética , Análisis de Secuencia de ARN , Técnicas de Movimiento Dental , Transcriptoma/genética , Proceso Alveolar , Animales , Regulación de la Expresión Génica , Masculino , Osteoclastos , Ratas , Microtomografía por Rayos X , Proteínas ras/metabolismo
15.
Angle Orthod ; 86(5): 782-8, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26927019

RESUMEN

OBJECTIVE: To evaluate and compare the effects of nanostructured, diamondlike, carbon (DLC) coating and nitrocarburizing on the frictional properties and biocompatibility of orthodontic stainless steel archwires. MATERIALS AND METHODS: Plasma-enhanced chemical vapor deposition technology was applied to coat DLC films onto the surface of austenitic stainless steel wires, and salt-bath nitrocarburizing technology was employed to achieve surface hardening of other wires. Surface and cross-sectional characteristics, microhardness, modulus of elasticity, friction resistance, corrosion resistance, and cell toxicity of the modified and control wires were analyzed. RESULTS: The surfaces of the DLC-coated and nitrocarburized wires were both smooth and even. Compared with the control, the DLC-coated wires were increased in surface hardness 1.46 times, decreased in elastic modulus, reduced in kinetic friction coefficient by 40.71%, and decreased in corrosion current density by two orders of magnitude. The nitrocarburized wire was increased in surface hardness 2.39 times, exhibited an unchanged elastic modulus, demonstrated a decrease in maximum static friction force of 22.2%, and rose in corrosion current density two orders of magnitude. Cytotoxicity tests revealed no significant toxicity associated with the modified wires. CONCLUSIONS: DLC coating and nitrocarburizing significantly improved the surface hardness of the wires, reduced friction, and exhibited good biocompatibility. The nanostructured DLC coating provided excellent corrosion resistance and good elasticity, and while the nitrocarburizing technique substantially improved frictional properties, it reduced the corrosion resistance of the stainless steel wires to a lesser extent.


Asunto(s)
Carbono , Aleaciones Dentales , Alambres para Ortodoncia , Estudios Transversales , Fricción , Ensayo de Materiales , Soportes Ortodóncicos , Acero Inoxidable , Propiedades de Superficie , Titanio
16.
Int J Biol Sci ; 12(10): 1155-1167, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27766031

RESUMEN

Osteoclasts originate from bone marrow monocyte/macrophage lineage cells, which are important for bone health. Class A scavenger receptor (SR-A) is a multifunctional molecule that functions during differentiation of monocyte into macrophages and osteoclasts. To further characterize the role of SR-A in osteoclasts, we used the murine tooth movement model (TM) and the murine anterior cruciate ligament transection model of osteoarthritis (ACLT OA). In these two models the bones involved are of different origin and have different properties. Bone resorption was decreased in SR-A-/- mice compared to SR-A+/+ mice. Further evaluation showed that the number of multinucleated osteoclasts in SR-A-/- mice, compared to SR-A+/+ mice, was significantly decreased both in vivo and in vitro. The levels of interleukin-6 (IL-6) produced by osteoclasts were reduced in SR-A-/- mice compared to SR-A+/+ mice. In the in vitro marrow-derived osteoclast formation assay and in both mouse models, osteoclastogenesis was restored to normal in SR-A-/- mice by administration of recombinant murine IL-6. Moreover, neutralization of IL-6 reduced the number of osteoclasts formed in SR-A+/+ mice of TM model. Both extracellular signal-regulated kinase (ERK) and c-Jun N-terminal protein kinase (JNK), but not p38, signaling pathways were downregulated in receptor activator of nuclear factor-κB ligand (RANKL)-stimulated SR-A-/- osteoclasts. Importantly, when treated with either ERK or JNK inhibitor, the numbers of osteoclasts generated from RANKL-induced bone marrow derived-macrophages of SR-A+/+ mice, and their IL-6 production, were significantly decreased. This suggests that SR-A activates the ERK and JNK signaling pathways, and promotes production of IL-6 by osteoclasts to further stimulate osteoclast formation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interleucina-6/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Osteoclastos/citología , Osteogénesis/fisiología , Receptores Depuradores de Clase A/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/genética , Interleucina-6/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Osteoartritis/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Ligando RANK/genética , Ligando RANK/metabolismo , Receptores Depuradores de Clase A/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA