Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur J Histochem ; 64(s2)2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33334091

RESUMEN

Our previous study documented that melatonin (MLT) induced the osteogenic differentiation of mesenchymal stem cells (MSCs) and promoted the healing of femoral fractures in rats via the neuropeptide Y (NPY)/neuropeptide Y1 receptor (NPY1R) signaling pathway. MLT treatment upregulated the expression of the zinc uptake transporter zinc transporter 1 (Zip1) in nerve cells. Prior research demonstrated that oral zinc upregulated NPY expression. MSCs were isolated from rat bone marrow and identified using flow cytometry in our study. The results showed that MLT treatment upregulated NPY and NPY1R levels in MSCs with osteogenic differentiation, which was accompanied by upregulated Zip1 expression. However, the MLT-induced osteogenic differentiation of MSCs was reversed after interference of Zip1 expression. It was confirmed by the decreased alkaline phosphatase (ALP) level; downregulated activities of type I collagen α1 chain (COL1A1), osteocalcin (OCN), runt-related transcription factor 2 (Runx2) and ALP; and reduced mineralized nodule formation. MLT promoted fracture healing in rats with femoral fracture, which was accompanied by increased expression of NPY and NPY1R and significantly increased expression of Zip1. In contrast, the silencing of Zip1 expression reversed MLT-mediated fracture healing. In summary, Zip1 participated in the regulation of the NPY/NPY1R signaling pathway via MLT to promote the osteogenic differentiation of MSCs and fracture healing.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Fracturas del Fémur/tratamiento farmacológico , Melatonina/uso terapéutico , Neuropéptido Y/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Fracturas del Fémur/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
2.
Cell Biol Int ; 44(2): 402-411, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31535749

RESUMEN

Melatonin has been reported to participate in bone metabolism in recent studies. However, the underlying mechanism in melatonin-mediated osteoblastic differentiation remains largely unknown. The aim of this study is to investigate the role of melatonin in osteoblastic differentiation. In the present study, additional melatonin significantly promoted osteoblastic differentiation of MC3T3-E1 cells as evidenced by increased messenger RNA (mRNA) levels of osteogenic markers, alkaline phosphatase (ALP), collagen type I α1 chain, osteocalcin, and runt-related transcription factor 2 (Runx2). It was noteworthy that the expression level of platelet-derived growth factor subunit B (PDGFB) and content of its homodimer PDGF-BB were remarkably increased after melatonin administration. Moreover, the mRNA levels of phosphorylated PDGFRß (PDGF receptor ß) and Akt, a serine/threonine-specific protein kinase, were significantly upregulated in melatonin-treated MC3T3-E1 cells determined by a real-time polymerase chain reaction. Besides, by performing alizarin red staining, osteoblastic differentiation of MC3T3-E1 cells was conspicuously promoted by melatonin, which could be partially attenuated by crenolanib, a PDGFR inhibitor. Similarly, results from immunofluorescence and western blot assay showed that melatonin-induced upregulation of Runx2 and phosphorylated Akt was suppressed by crenolanib. Akt inhibition by MK-2206 also suppressed osteoblastic differentiation. Furthermore, by in vivo assay, additional melatonin promoted osteoblastic differentiation in mice with femoral fracture, and obvious callus formation was observed in melatonin-treated mice 5 weeks after fracture. Melatonin supplement also inhibited osteoclastic differentiation in mice. All statistical analysis was performed using GraphPad Prism and a P < 0.05 was deemed to be significant. To summarize, we demonstrate that melatonin promotes osteoblastic differentiation in MC3T3-E1 cells and enhances fracture healing in mouse femoral fracture model and regulates PDGF/AKT signaling pathway.


Asunto(s)
Antioxidantes/farmacología , Diferenciación Celular , Melatonina/farmacología , Osteoblastos/citología , Osteogénesis , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal
3.
J Int Med Res ; 48(3): 300060519883731, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31852342

RESUMEN

OBJECTIVE: MicroRNAs (miRNAs) are reported to have crucial roles in human cancers; however, their role in colorectal cancer (CRC) remains largely unknown. METHODS: In this study, we analyzed the expression of miR-4324 in CRC cell lines using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). We also examined miR-4324 expression in CRC tumor tissues using a miRNA expression dataset obtained from the Gene Expression Omnibus. We validated the connection between miR-4324 and homeobox B2 (HOXB2) using a luciferase activity reporter assay and western blotting. The effects of miR-4324 and HOXB2 on CRC cell malignant behaviors in vitro were further investigated. RESULTS: miR-4324 expression was significantly decreased in both CRC tumor tissues and cell lines. Overexpression of miR-4324 suppressed CRC cell proliferation, migration, and invasion. In contrast, overexpression of HOXB2 promoted CRC malignant cell behaviors. Furthermore, we validated HOXB2 as a direct target of miR-4324. CONCLUSIONS: miR-4324 expression was decreased in CRC. miR-4324 regulates CRC cell proliferation, migration, and invasion by targeting HOXB2.


Asunto(s)
Neoplasias Colorrectales , Proteínas de Homeodominio , MicroARNs , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Humanos , MicroARNs/genética , Invasividad Neoplásica/genética , Factores de Transcripción
4.
Pharmacology ; 102(5-6): 272-280, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30227410

RESUMEN

The function of melatonin (MLT) in promoting fracture healing has been demonstrated in previous studies. However, the molecular mechanism underlying therapeutic effects of MLT is not entirely clear. In this study, mesenchymal stem cells (MSCs) were isolated from rat bone marrow and identified by flow cytometry. We found that MLT treatment upregulated the neuropeptide Y (NPY) and NPY receptor Y1 (NPY1R) expression, and promoted the proliferation and migration of MSCs, which was suppressed by BIBP3226, an inhibitor of NPY1R. Moreover, the levels of NPY and NPY1R in MSCs undergoing osteoblastic differentiation were upregulated after MLT administration. MLT-induced osteoblastic differentiation of MSCs was suppressed by BIBP3226 treatment, as evidenced by decreased levels of alkaline phosphatase (ALP), collagen type I α1 chain, osteocalcin, and runt-related transcription factor 2, downregulated activity of ALP, as well as reduced calcium nodule formation. Furthermore, we demonstrated that MLT could promote fracture healing in a rat model of femoral fracture, which was accompanied by the elevated expression of NPY and NPY1R. The administration of BIBP3226 inhibited fracture healing mediated by MLT. To sum up, our results show that MLT promotes osteoblastic differentiation of MSCs and fracture healing by NPY/NPY1R signaling.


Asunto(s)
Curación de Fractura/efectos de los fármacos , Melatonina/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Neuropéptido Y/metabolismo , Osteoblastos/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Diferenciación Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Fracturas del Fémur/tratamiento farmacológico , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
Oncotarget ; 8(51): 88599-88612, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29179460

RESUMEN

Zinc finger protein 281 (ZNF281) has been recently shown to be critical for CRC progression. However, the immediate upstream regulators of ZNF281 remain unclear. Here we reported that the E3 ligase the ß-transducin repeat-containing protein 2 (ß-TrCP2) governs the ubiquitination and degradation of ZNF281. In human CRC specimens, endogenous ß-TrCP2 were inversely correlated with ZNF281. Beta-TrCP2 reversed the phenotype of CRC cell with overexpressed ZNF281. Moreover, we found that glycogen synthase kinase 3ß (GSK-3ß), not GSK-α, could bind to and phosphorylate ZNF281 at one consensus motif (TSGEHS; phosphorylation site is shown in italics), which promotes the interaction of ZNF281 with ß-TrCP2, not ß-TrCP1, and leads to the subsequent ubiquitination and degradation of phosphorylated ZNF281. A mutant of ZNF281 (ZNF281-S638A) is much more stable than wild-type ZNF281 because ZNF281-S638A mutant abolishes the phosphorylation by GSK-3ß and can not be ubiquitinated and degraded by ß-TrCP2. Conversely, ZNF281 transcriptionally repressed the expression of ß-TrCP2, indicating a negative feedback loop between ZNF281 and ß-TrCP2 in CRC cells. These findings suggest that the turnover of ZNF281 by ß-TrCP2 might provide a potentially novel treatment for patients with CRC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...