Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Stem Cells ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38864549

RESUMEN

SIRT6 owns versatile types of enzymatic activities as a multitasking protein, including ribosyltransferase and deacetylase ones. To investigate the epigenetic regulations of SIRT6 on MSC fate determination via histone deacetylation, we utilized allosteric small molecules specifically controlling its histone 3 deacetylation activities. Results showed that enhanced deacetylation of SIRT6 promoted the ossific lineage commitment of MSC and finally achieved anabolic effects on hard tissues. Mechanistically, H3K9ac and H3K56ac, governed by SIRT6, in MSC orchestrated the transcriptions of crucial metabolic genes, mediating MSC fate determination. Most importantly, our data evidenced that modulating the epigenetic regulations of SIRT6, specifically via enhancing its deacetylation of H3K9ac and H3K56ac, was a promising choice to treat bone loss diseases and promote dentine regeneration. In this study, we revealed the specific roles of SIRT6's histone modification in MSC fate determination. These findings endow us with insights on SIRT6 and the promising therapeutic choices through SIRT6's epigenetic functions for hard tissues regeneration.

2.
FASEB J ; 36(6): e22377, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35608871

RESUMEN

Osteoarthritis (OA) is the leading joint disease characterized by cartilage destruction and loss of mobility. Accumulating evidence indicates that the incidence and severity of OA increases with diabetes, implicating systemic glucose metabolism in joint health. However, a definitive link between cellular metabolism in articular cartilage and OA pathogenesis is not yet established. Here, we report that in mice surgically induced to develop knee OA through destabilization of medial meniscus (DMM), expression of the main glucose transporter Glut1 is notably reduced in joint cartilage. Inducible deletion of Glut1 specifically in the Prg4-expressing articular cartilage accelerates cartilage loss in DMM-induced OA. Conversely, forced expression of Glut1 protects against cartilage destruction following DMM. Moreover, in mice with type I diabetes, both Glut1 expression and the rate of glycolysis are diminished in the articular cartilage, and the diabetic mice exhibit more severe cartilage destruction than their nondiabetic counterparts following DMM. The results provide proof of concept that boosting glucose metabolism in articular chondrocytes may ameliorate cartilage degeneration in OA.


Asunto(s)
Cartílago Articular , Diabetes Mellitus Experimental , Osteoartritis , Animales , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Ratones , Osteoartritis/metabolismo
3.
J Am Chem Soc ; 144(22): 9926-9937, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35616998

RESUMEN

The development of lipid nanoparticle (LNP) formulations for targeting the bone microenvironment holds significant potential for nucleic acid therapeutic applications including bone regeneration, cancer, and hematopoietic stem cell therapies. However, therapeutic delivery to bone remains a significant challenge due to several biological barriers, such as low blood flow in bone, blood-bone marrow barriers, and low affinity between drugs and bone minerals, which leads to unfavorable therapeutic dosages in the bone microenvironment. Here, we construct a series of bisphosphonate (BP) lipid-like materials possessing a high affinity for bone minerals, as a means to overcome biological barriers to deliver mRNA therapeutics efficiently to the bone microenvironment in vivo. Following in vitro screening of BP lipid-like materials formulated into LNPs, we identified a lead BP-LNP formulation, 490BP-C14, with enhanced mRNA expression and localization in the bone microenvironment of mice in vivo compared to 490-C14 LNPs in the absence of BPs. Moreover, BP-LNPs enhanced mRNA delivery and secretion of therapeutic bone morphogenetic protein-2 from the bone microenvironment upon intravenous administration. These results demonstrate the potential of BP-LNPs for delivery to the bone microenvironment, which could potentially be utilized for a range of mRNA therapeutic applications including regenerative medicine, protein replacement, and gene editing therapies.


Asunto(s)
Lípidos , Nanopartículas , Animales , Difosfonatos/farmacología , Liposomas , Ratones , ARN Mensajero/genética , ARN Interferente Pequeño/genética
4.
Cancer Sci ; 112(10): 4139-4150, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34058054

RESUMEN

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) (eg, gefitinib) exert potent therapeutic efficacy in non-small-cell lung cancer (NSCLC) harboring EGFR-activating mutations. However, the resistance to EGFR TKIs limits their clinical therapeutic efficacy. TIP30, a newly identified tumor suppressor, appears to be involved in the regulation of cytoplasmic and nuclear EGFR signaling in NSCLC. Our previous study demonstrated that TIP30 regulated EGF-dependent cyclin D1 transcription in human lung adenocarcinoma and suppressed tumorigenesis. In the present study, the involvement of TIP30 in combating gefitinib resistance in NSCLC was determined for the first time in vitro and in vivo. Gain and loss of function studies showed that overexpression of TIP30 effectively sensitized cells to gefitinib in vitro, whereas TIP30 inhibition promoted gefitinib cell resistance. Moreover, TIP30 negatively regulated the activation of the p-AKT and p-MEK signaling pathways in PC9/GR. Importantly, PC9/GR harbored high levels of nuclear EGFR, and overexpression of TIP30 restored irregular EGFR trafficking and degradation from early endosomes to the late endosomes, decreasing the nuclear accumulation of EGFR, which may partly or totally inhibit EGFR-mediated induction of c-Myc transcription. Xenographic tumors induced by overexpression of TIP30 by PC9/GR cells in nude mice were suppressed compared with their original counterparts. Overall, it was revealed that TIP30 overexpression restored gefitinib sensitivity in NSCLC cells and attenuated the cytoplasmic and nuclear EGFR signaling pathways and may be a promising biomarker in gefitinib resistance in NSCLC.


Asunto(s)
Acetiltransferasas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Resistencia a Antineoplásicos/fisiología , Neoplasias Pulmonares/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Núcleo Celular/metabolismo , Ciclina D1/metabolismo , Citoplasma/metabolismo , Endosomas/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinib/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Lisosomas/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Stem Cells ; 38(8): 1020-1033, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32346881

RESUMEN

As a profoundly anabolic regulator of bone, Wnt7b is well acknowledged to enhance osteoblast activities. Here, we report that bone marrow mesenchymal stem cells (BMSCs) are another important population responding to Wnt7b. In this study, we systematically investigated the in vivo role of Wnt7b in BMSCs using transgenic mice, high-throughput RNA-seq, immunohistochemistry, RT-qPCR, and in situ hybridization. These methods led us to uncover that Sox11 is induced via Wnt7b in BMSCs. Colony formation assay, flow cytometry, EdU incorporation labeling, RT-qPCR, and Western blot were conducted to detect the self-renewal capacity of BMSCs. Alkaline phosphatase staining, alizarin red staining, and ex vivo BMSCs transplantation were utilized to detect the osteogenic ability of BMSCs. ChIP-qPCR, shRNAs, and immunofluorescence staining were utilized to investigate the underlying mechanisms. Consequently, bone-derived Wnt7b was found to decrease in osteoporosis and elevate in bone fracture healing. During bone fracture healing, Wnt7b was particularly expressed in the mesenchymal cells residing within healing frontiers. RNA-seq data of Wnt7b-overexpressed bones uncovered the significant upregulation of Sox11. Histological results further unveiled that Sox11 is specifically increased in BMSCs. Wnt7b-induced Sox11 was demonstrated to reinforce both self-renewal and osteogenic differentiation of BMSCs. Mechanistically, Wnt7b activates the Ca2+ -dependent Nfatc1 signaling to directly induce Sox11 transcription, which in turn activates the transcriptions of both proliferation-related transcription factors (Ccnb1 and Sox2) and osteogenesis-related factors (Runx2, Sp7) in BMSCs. It is intriguing that this Wnt7b-Sox11 signaling in BMSCs is ß-Catenin-independent. Overall, this study provides brand new insights of Wnt7b in bone formation, namely, Wnt7b can enhance both self-renewal and osteogenic differentiation of BMSCs via inducing Sox11. These findings present a new crosstalk between Wnt and Sox signaling in BMSCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción SOXC/metabolismo , Proteínas Wnt/metabolismo , Animales , Células de la Médula Ósea/citología , Modelos Animales de Enfermedad , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal
6.
bioRxiv ; 2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36711657

RESUMEN

Skeletal fragility is associated with type 2 diabetes mellitus (T2D), but the underlying mechanism is not well understood. Here, in a mouse model for youth-onset T2D, we show that both trabecular and cortical bone mass are reduced due to diminished osteoblast activity. Stable isotope tracing in vivo with 13 C-glucose demonstrates that both glycolysis and glucose fueling of the TCA cycle are impaired in diabetic bones. Similarly, Seahorse assays show suppression of both glycolysis and oxidative phosphorylation by diabetes in bone marrow mesenchymal cells as a whole, whereas single-cell RNA sequencing reveals distinct modes of metabolic dysregulation among the subpopulations. Metformin not only promotes glycolysis and osteoblast differentiation in vitro, but also improves bone mass in diabetic mice. Finally, targeted overexpression of Hif1a or Pfkfb3 in osteoblasts of T2D mice averts bone loss. The study identifies osteoblast-intrinsic defects in glucose metabolism as an underlying cause of diabetic osteopenia, which may be targeted therapeutically.

7.
Elife ; 122023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37144869

RESUMEN

Skeletal fragility is associated with type 2 diabetes mellitus (T2D), but the underlying mechanism is not well understood. Here, in a mouse model for youth-onset T2D, we show that both trabecular and cortical bone mass is reduced due to diminished osteoblast activity. Stable isotope tracing in vivo with 13C-glucose demonstrates that both glycolysis and glucose fueling of the TCA cycle are impaired in diabetic bones. Similarly, Seahorse assays show suppression of both glycolysis and oxidative phosphorylation by diabetes in bone marrow mesenchymal cells as a whole, whereas single-cell RNA sequencing reveals distinct modes of metabolic dysregulation among the subpopulations. Metformin not only promotes glycolysis and osteoblast differentiation in vitro, but also improves bone mass in diabetic mice. Finally, osteoblast-specific overexpression of either Hif1a, a general inducer of glycolysis, or Pfkfb3 which stimulates a specific step in glycolysis, averts bone loss in T2D mice. The study identifies osteoblast-intrinsic defects in glucose metabolism as an underlying cause of diabetic osteopenia, which may be targeted therapeutically.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Ratones , Masculino , Animales , Osteogénesis , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Experimental/metabolismo , Osteoblastos/metabolismo , Glucosa/metabolismo
8.
JCI Insight ; 8(24)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-37917194

RESUMEN

Osteoclasts specialize in bone resorption and are critical for bone remodeling. Previous studies have shown that osteoclasts possess abundant mitochondria and derive most energy through oxidative phosphorylation (OXPHOS). However, the energy substrates fueling OXPHOS in osteoclasts remain to be fully defined. Here, we showed that osteoclast differentiation was coupled with increased oxidation of glucose, glutamine, and oleate. Transcriptomic analyses with RNA sequencing revealed marked upregulation of genes participating in OXPHOS and mitochondrial fatty acid oxidation, during osteoclast differentiation. Increased mitochondrial oxidation of long-chain fatty acids was required for osteoclast differentiation in vitro. However, blocking fatty acid oxidation in vivo, by deletion of carnitine palmitoyltransferase 1a (Cpt1a) in osteoclast progenitors, impaired osteoclast formation only in the female mice. The Cpt1a-deficient females were further protected from osteoclast activation by a high-fat diet. The males, on the contrary, exhibited normal bone resorption despite Cpt1a deletion, regardless of the dietary fat content. Moreover, concurrent deletion of mitochondrial pyruvate carrier 1 and Cpt1a, blocking mitochondrial oxidation of both glucose and fatty acids in the osteoclast lineage, failed to impede bone resorption in the males. The study therefore uncovers a female-specific dependence on mitochondrial oxidation of fatty acids and glucose in osteoclasts in vivo.


Asunto(s)
Resorción Ósea , Osteoclastos , Masculino , Ratones , Femenino , Animales , Osteoclastos/metabolismo , Caracteres Sexuales , Resorción Ósea/metabolismo , Mitocondrias , Ácidos Grasos/metabolismo , Glucosa/metabolismo
9.
Cell Chem Biol ; 30(9): 1053-1063.e5, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37562406

RESUMEN

Type I diabetes (T1D) impairs bone accrual in patients, but the mechanism is unclear. Here in a murine monogenic model for T1D, we demonstrate that diabetes suppresses bone formation resulting in a rapid loss of both cortical and trabecular bone. Single-cell RNA sequencing uncovers metabolic dysregulation in bone marrow osteogenic cells of diabetic mice. In vivo stable isotope tracing reveals impaired glycolysis in diabetic bone that is highly responsive to insulin stimulation. Remarkably, deletion of the insulin receptor reduces cortical but not trabecular bone. Increasing glucose uptake by overexpressing Glut1 in osteoblasts exacerbates bone defects in T1D mice. Conversely, activation of glycolysis by Pfkfb3 overexpression preserves both trabecular and cortical bone mass in the face of diabetes. The study identifies defective glucose metabolism in osteoblasts as a pathogenic mechanism for osteopenia in T1D, and furthermore implicates boosting osteoblast glycolysis as a potential bone anabolic therapy.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Humanos , Ratones , Animales , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Osteoblastos/metabolismo , Densidad Ósea , Glucólisis
10.
Cell Rep ; 36(7): 109542, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34407400

RESUMEN

Teriparatide is the most widely prescribed bone anabolic drug in the world, but its cellular targets remain incompletely defined. The Gli1+ metaphyseal mesenchymal progenitors (MMPs) are a main source for osteoblasts in postnatal growing mice, but their potential response to teriparatide is unknown. Here, by lineage tracing, we show that teriparatide stimulates both proliferation and osteoblast differentiation of MMPs. Single-cell RNA sequencing reveals heterogeneity among MMPs, including an unexpected chondrocyte-like osteoprogenitor (COP). COP expresses the highest level of Hedgehog (Hh) target genes and the insulin-like growth factor 1 receptor (Igf1r) among all cell clusters. COP also expresses Pth1r and further upregulates Igf1r upon teriparatide treatment. Inhibition of Hh signaling or deletion of Igf1r from MMPs diminishes the proliferative and osteogenic effects of teriparatide. The study therefore identifies COP as a teriparatide target wherein Hh and insulin-like growth factor (Igf) signaling are critical for the osteoanabolic response in growing mice.


Asunto(s)
Huesos/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Somatomedinas/metabolismo , Células Madre/metabolismo , Teriparatido/farmacología , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Huesos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Mesodermo/citología , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , RNA-Seq , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos
11.
Arch Oral Biol ; 104: 123-132, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31181411

RESUMEN

OBJECTIVES: This study investigates the role of Wnt7b in mouse dentin formation. DESIGN: C57BL/6 mouse tooth germs at different developmental stages were collected to measure the expression of Wnt7b by immunohistochemical staining. The morphology of mandibles of Dmp1-cre;ROSA26-Wnt7b transgenic mice and ROSA26-Wnt7b littermates was analyzed by Micro-CT and HE staining. The ultramicrostructure of dentin was scanned with an electron microscope. Primary mouse dental papillae cells (MDPCs) and odontoblastic cell line (A11) were cultured and infected with adenovirus to overexpress Wnt7b. Cell proliferation and cell apoptosis were evaluated using CCK-8 and flow cytometry. Osteogenic differentiation of MDPCs and A11 was assessed by Alizarin red staining, and qPCR detection of osteogenic gene expression. The activation of signaling pathways was measured by the use of western blot analysis. The ERK1/2 inhibitor was used to test the effect of Wnt7b regulated cell differentiation. RESULTS: Wnt7b was expressed principally in the mouse odontoblast layer after the early bell stage. In transgenic mice, Wnt7b was over-expressed in tooth mesenchyme, with a thinner predentin layer and thicker intertubular dentin. Both the micro-hardness value and the Ca/Pi ratio of dentin of transgenic mice were higher. Wnt7b promoted proliferation and mineralization of MDPCs and A11. The protein level of p-ERK1/2 was found to be higher in A11 infected with Ad-Wnt7b. The ERK signaling pathway inhibitor partly rescued the Wnt7b-induced differentiation of A11. CONCLUSIONS: Wnt7b enhances dentinogenesis by increasing the proliferation and differentiation of dental mesenchymal cells partly through ERK1/2 pathway.


Asunto(s)
Dentinogénesis , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas , Proteínas Wnt , Animales , Diferenciación Celular , Pulpa Dental , Dentinogénesis/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Odontoblastos , Osteogénesis , Proteínas Proto-Oncogénicas/fisiología , Proteínas Wnt/fisiología
12.
Sci Rep ; 9(1): 8073, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31147568

RESUMEN

With the rapid development of tissue engineering therapies, there is a growing interest in the application of recombinant human growth factors (rhGFs) to regenerate human orofacial bones. However, despite reports of their ability to promote orofacial bone regeneration in animal experiments, their benefits in human clinical treatments remain unclear. Furthermore, the appropriate concentrations or indications of a specific rhGF remain ambiguous. Therefore it is essential to collect data from diverse clinical trials to evaluate their effects more precisely. Here we reviewed randomized clinical trials (RCT) that focused on the utilization of rhGFs in orofacial bone regeneration. Data from included studies were extracted, pooled and then quantitatively analyzed according to a pre-established protocol. Our results indicate that all current concentrations of rhBMP-2 produces insufficient effect on promoting either tooth extraction socket healing, sinus augmentation or reconstruction of alveolar clefts. However, 0.3 mg/ml rhPDGF-BB promotes the healing of tooth extraction sockets, though the effect does not reach a level of statistical significance. Summarily, we recommend concentrations of 0.3 mg/ml rhPDGF-BB only for the healing of tooth extraction sockets.


Asunto(s)
Becaplermina/administración & dosificación , Proteína Morfogenética Ósea 2/administración & dosificación , Regeneración Ósea/efectos de los fármacos , Huesos Faciales/efectos de los fármacos , Factor de Crecimiento Transformador beta/administración & dosificación , Injerto de Hueso Alveolar/métodos , Relación Dosis-Respuesta a Droga , Huesos Faciales/fisiología , Huesos Faciales/cirugía , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Recombinantes/administración & dosificación , Elevación del Piso del Seno Maxilar/métodos , Resultado del Tratamiento , Cicatrización de Heridas/efectos de los fármacos
13.
Sci Rep ; 8(1): 10184, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29977065

RESUMEN

Osteoclasts are multinucleated giant cells. Fusion is an essential element in the formation of osteoclasts. However, the exact cellular events and mechanisms remain largely unknown because of limited and insufficient methods for observing fusion process. In this work, a fluorescence reporter strategy was established to monitor osteoclast fusion. After fusing with cells expressing Cre recombinase, those cells with double fluorescence switch its expression from red to green fluorescent protein. The effect of RANKL and PTH on osteoclast fusion were both quantitatively and visually detected utilizing this strategy. Furthermore, a combination of this strategy with a technique of fluorescence-activated cell sorting revealed two different populations of fused osteoclasts, tdTomato+ GFP+ cells (TG cells) and GFP+ cells (G cells). The results argue for the potential of combining this technique with other bio-technologies to gain more information about osteoclast fusion. Overall, these data demonstrated that this visual fluorescence switch strategy is useful for further analysis of osteoclast fusion mechanisms.


Asunto(s)
Microscopía Intravital/métodos , Osteoclastos/fisiología , Animales , Fusión Celular , Separación Celular/métodos , Citometría de Flujo/métodos , Colorantes Fluorescentes/química , Genes Reporteros/genética , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Ratones , Microscopía Fluorescente/métodos , Osteoclastos/efectos de los fármacos , Cultivo Primario de Células , Células RAW 264.7
14.
PLoS One ; 12(3): e0174255, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28355287

RESUMEN

Dental Mesenchymal Cells (DMCs) are known to play a role in tooth development as well as in the repair and regeneration of dental tissue. A large number of signaling molecules regulate the proliferation and differentiation of DMC, though the underlying mechanisms are still not fully understood. Sirtuin-6 (SIRT6), a key regulator of aging, can exert an impact on embryonic stem cell (ESC) differentiation. The experimental deletion of Sirt6 in mouse bone marrow cells has been found to have an inhibiting impact on the bone mineral density and the osteogenic differentiation of these cells. The possible role of Sirt6 in tooth development, however, has at present remained largely unexplored. In the present study, we found that SIRT6 had no effect on tooth development before birth. However, Sirt6 gene deletion in knockout mice did have two post-natal impacts: a delay in tooth eruption and sluggishness in the development of dental roots. We propose an explanation of the possible molecular basis of the changes observed in Sirt6-/- mice. SIRT6 is expressed in mouse odontoblasts. Sirt6 deletion enhanced the proliferation of DMCs, as well as their capacity for adipogenic differentiation. On the other hand, it inhibited their capacity for in vitro osteogenic/chondrogenic differentiation. Further studies suggested that other factors may mediate the role of Sirt6 in odontogenesis. These include the nuclear factor kappa B (NF-κB), p38 mitogen-activated protein kinase (p38-MAPK), extracellular regulated MAP kinase (ERK) pathways and the mitochondrial energy. We demonstrated that Sirt6 plays a role in tooth root formation and confirmed that SIRT6 is necessary for DMC differentiation as well as for the development of the tooth root and for eventual tooth eruption. These results establish a new link between SIRT6 and tooth development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Diente Molar/metabolismo , Odontoblastos/metabolismo , Sirtuinas/genética , Raíz del Diente/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular , Embrión de Mamíferos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Diente Molar/crecimiento & desarrollo , FN-kappa B/genética , FN-kappa B/metabolismo , Odontoblastos/citología , Odontogénesis/genética , Transducción de Señal , Sirtuinas/deficiencia , Erupción Dental/genética , Raíz del Diente/crecimiento & desarrollo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA