Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Med ; 28(1): 134, 2022 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-36401176

RESUMO

BACKGROUND: Circular RNA (circ) AFF4 was documented to regulate osteogenesis but the underlying mechanism remains to be elucidated. The preliminary study showed that circ_AFF4 may promote osteogenesis via FNDC5/Irisin. Furthermore, the online prediction tool indicated the interaction of circ_AFF4, insulin-like growth factor-2 mRNA-binding protein 3 (IGF2BP3), FNDC5 and lysine (K)-specific demethylase 1 A (KDM1A). Therefore, this study aims to elucidate the relationships of KDM1A, circ_AFF4, IGF2BP3 and FNDC5/Irisin during osteogenesis. METHODS: The alkaline phosphatase (ALP) activities and osteogenic-related factors were determined using ALP and alizarin red S (ARS) staining, real-time quantitative PCR(RT-qPCR) and western blot. Immunoprecipitation (RIP), pull-down assay and fluorescence in situ hybridization (FISH) were used to examine the interactions among circ_AFF4/IGF2BP3/FNDC5. A mouse in vivo model was utilized to further confirm the regulatory effect on bone formation. RESULTS: Circ_AFF4 and KDM1A expression levels were increased during osteoinduction of BM-MSCs. Knockdown of circ_AFF4 and KDM1A significantly suppressed BM-MSC osteogenesis. We also proved that KDM1A directly bound to circ_AFF4 and FNDC5 promoter and induced circ_AFF4 and FNDC5 expression. Furthermore, circ_AFF4 enhanced the stability of FNDC5 by generating a circ_AFF4, IGF2BP3 and FNDC5 RNA-protein complex, and thereby induced Irisin and osteogenesis. The in vitro data was confirmed with in vivo model. CONCLUSION: These findings elucidate that KDM1A induces circ_AFF4, which promotes promote osteogenesis via IGF2BP3. This study indicates that circ_AFF4 may potentially represent a critical therapeutic target for the diseases.


Assuntos
Osteogênese , RNA Circular , Camundongos , Animais , RNA Circular/genética , Osteogênese/genética , Fibronectinas/genética , Hibridização in Situ Fluorescente , Fatores de Transcrição/genética
2.
Cell Death Dis ; 12(7): 631, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145212

RESUMO

Bone marrow-derived mesenchymal stem cells (BM-MSCs), the common progenitor cells of adipocytes and osteoblasts, have been recognized as the key mediator during bone formation. Herein, our study aim to investigate molecular mechanisms underlying circular RNA (circRNA) AFF4 (circ_AFF4)-regulated BM-MSCs osteogenesis. BM-MSCs were characterized by FACS, ARS, and ALP staining. Expression patterns of circ_AFF4, miR-135a-5p, FNDC5/Irisin, SMAD1/5, and osteogenesis markers, including ALP, BMP4, RUNX2, Spp1, and Colla1 were detected by qRT-PCR, western blot, or immunofluorescence staining, respectively. Interactions between circ_AFF4 and miR-135a-5p, FNDC5, and miR-135a-5p were analyzed using web tools including TargetScan, miRanda, and miRDB, and further confirmed by luciferase reporter assay and RNA pull-down. Complex formation between Irisin and Integrin αV was verified by Co-immunoprecipitation. To further verify the functional role of circ_AFF4 in vivo during bone formation, we conducted animal experiments harboring circ_AFF4 knockdown, and born samples were evaluated by immunohistochemistry, hematoxylin and eosin, and Masson staining. Circ_AFF4 was upregulated upon osteogenic differentiation induction in BM-MSCs, and miR-135a-5p expression declined as differentiation proceeds. Circ_AFF4 knockdown significantly inhibited osteogenesis potential in BM-MSCs. Circ_AFF4 stimulated FNDC5/Irisin expression through complementary binding to its downstream target molecule miR-135a-5p. Irisin formed an intermolecular complex with Integrin αV and activated the SMAD1/5 pathway during osteogenic differentiation. Our work revealed that circ_AFF4, acting as a sponge of miR-135a-5p, triggers the promotion of FNDC5/Irisin via activating the SMAD1/5 pathway to induce osteogenic differentiation in BM-MSCs. These findings gained a deeper insight into the circRNA-miRNA regulatory system in the bone marrow microenvironment and may improve our understanding of bone formation-related diseases at physiological and pathological levels.


Assuntos
Células da Medula Óssea/metabolismo , Diferenciação Celular , Fibronectinas/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Osteogênese , RNA Circular/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Animais , Células Cultivadas , Feminino , Fibronectinas/genética , Humanos , Transplante de Células-Tronco Mesenquimais , Camundongos Endogâmicos BALB C , MicroRNAs/genética , RNA Circular/genética , Transdução de Sinais
3.
J Orthop Res ; 38(3): 670-679, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31608495

RESUMO

Lumbar facet joint osteoarthritis (LFJ OA) is regarded as one of the common causes of low back pain (LBP). The pathogenesis and underlying mechanism of this disease are largely unknown, there is still no effective disease-modifying therapy. This study aims to investigate the efficacy of exosomes derived from bone marrow mesenchymal stem cells (BMSCs) on the pathogenesis and behavioral signs of LBP in the LFJ OA mouse model. The pathogenetic change in cartilage and aberrant nerve invasion in the subchondral bone of LFJ in a mouse model after treatment with BMSC-exosomes was evaluated. BMSC-exosomes could relieve pain via abrogation of aberrant CGRP-positive nerve and abnormal H-type vessel formation in the subchondral bone of LFJ. Moreover, BMSC-exosomes attenuated cartilage degeneration and inhibited tartrate-resistant acid phosphatase expression and RANKL-RANK-TRAF6 signaling activation to facilitate subchondral bone remodeling. These results indicated that BMSC-exosomes could relive behavioral signs of LBP and pathological processes in LFJ OA. BMSC-exosomes have a prominent protective effect and might be a potential therapeutic option for the treatment of LFJ OA causing LBP. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:670-679, 2020.


Assuntos
Exossomos/metabolismo , Vértebras Lombares/patologia , Células-Tronco Mesenquimais/metabolismo , Osteoartrite/terapia , Manejo da Dor/métodos , Animais , Células da Medula Óssea/citologia , Remodelação Óssea , Cartilagem Articular/inervação , Cartilagem Articular/patologia , Processamento de Imagem Assistida por Computador/métodos , Imageamento Tridimensional , Vértebras Lombares/inervação , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/patologia , Ligante RANK/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo
4.
Onco Targets Ther ; 12: 11667-11677, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32021245

RESUMO

BACKGROUND: Osteosarcoma (OS) is one of the most difficult cancers to treat due to its resistance to chemotherapy. The essential role played by Mcl-1 in promoting chemoresistance has been observed in a variety of cancers, including OS, while the underlying mechanism remains unclear. METHODS: We investigated the expression of Mcl-1 in 42 paired OS specimens obtained before and after adjuvant chemotherapy, and its correlation with clinicopathological characteristics. Loss and gain of function studies were performed to analyze the effects of Mcl-1 modulations on the chemosensitivity, and the mechanism involved in the deregulation of Mcl-1 in OS cells. RESULTS: In OS specimens, the expression of Mcl-1 was significantly upregulated after chemotherapy, and high Mcl-1 expression was associated with poorer overall survival and an increased recurrence rate. Furthermore, we demonstrated that chemotherapy-driven increased Mcl-1 decreased chemosensitivity by promoting tumour proliferation and inhibiting DNA damage. Moreover, Mcl-1 was found to be a direct target of miR-375 in OS cells. The knockdown of Mcl-1 phenocopied miR-375 downregulation, and the overexpression of miR-375 rescued the effects of cisplatin-induced DNA damage mediated by Mcl-1. CONCLUSION: Our data indicated that chemotherapy-driven increase in the expression of Mcl-1 plays a critical role in chemoresistance, and the intervention of the miR-375/Mcl-1 axis may offer a novel strategy to enhance chemosensitivity in OS treatment.

5.
Exp Ther Med ; 8(5): 1381-1388, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25289027

RESUMO

Tricalcium phosphate (TCP) and platelet-rich plasma (PRP) are commonly used in bone tissue engineering. The aim of the present study was to investigate a composite that combined TCP with PRP and assess its effectiveness in the treatment of bone defects. Cavity-shaped bone defects were established on the tibiae of 27 beagle dogs, and were repaired by pure ß-TCP with bone marrow stromal cells (BMSCs), ß-TCP/PRP with BMSCs and autogenic ilium. The samples were harvested at 4, 8 and 12 weeks, and bone regeneration was evaluated using X-ray radiography, immunocytochemical staining of osteocalcin (OCN), hematoxylin and eosin staining and reverse transcription-polymerase chain reaction analyses. Biomechanical tests of the scaffolds were performed at the 12th week after scaffold implantation. When using pure ß-TCP as a scaffold, the scaffold-bone interface was clear and no material adsorption and bone healing was observed. Substantial bone regeneration was observed when the tibial defects were restored using ß-TCP/PRP and autogenic ilium. Furthermore, the mRNA expression levels of OCN, alkaline phosphatase and collagen type I α1 were significantly higher in the animals with ß-TCP/PRP scaffolds at 8 and 12 weeks following implantation compared with those in the animals with the pure ß-TCP scaffolds. The maximum load and compressive strength of the ß-TCP/PRP scaffolds were similar to those of the autogenic ilium; however, they were significantly higher than those of the pure ß-TCP scaffold. Thus, the ß-TCP/PRP composite may be used as a potential scaffold to carry in vitro cultured BMSCs to treat bone defects.

6.
Exp Ther Med ; 8(4): 1075-1080, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25187799

RESUMO

Whether the levels of procalcitonin (PCT) in the serum and synovial fluid are effective indicators for distinguishing septic arthritis (SA) from non-infectious arthritis remains controversial. The present study aimed to evaluate whether PCT levels in fresh serum or fresh joint fluid may be used in the differential diagnosis of SA from rheumatoid arthritis (RA), osteoarthritis (OA) and gouty arthritis (GA). From January 2012 to June 2013, 23 patients with knee SA, 21 patients with RA, 40 patients with OA and 11 patients with GA were enrolled in the current study. The levels of PCT were measured within 24 h after specimen collection at room temperature. An enzyme-linked fluorescence assay (ELFA) was used to detect the levels of PCT in the serum and synovial fluid. The correlations between the levels of PCT in the serum and synovial fluid and the arthritic patient groups were determined by the Nemenyi test. Areas under the receiver operating characteristic (ROC) curve were calculated to evaluate the accuracy of the correlations. The levels of PCT in the serum and joint fluid of the patients in the SA group were higher compared with those of the other groups (P<0.01) and there were no significant differences among the RA, OA and GA groups in these levels. A PCT level of <0.5 µg/l in the serum and synovial fluid had high specificity in the differential diagnosis of SA from RA, OA and GA. Synovial fluid PCT revealed significantly greater sensitivity than serum PCT. The accuracy of the differential diagnosis of SA by the serum levels of PCT was significantly lower than that by the synovial fluid levels of PCT. The levels of PCT in the serum and synovial fluid may be used as alternative laboratory indicators to distinguish between SA and the non-infectious types of arthritis; however, the PCT levels in fresh synovial fluid are more sensitive and accurate indicators than PCT levels in fresh serum.

7.
Exp Ther Med ; 8(4): 1081-1086, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25187800

RESUMO

ß-tricalcium phosphate (ß-TCP) and platelet-rich plasma (PRP) are commonly used in bone tissue engineering. In the present study, a new composite combining strengthened ß-TCP and PRP was prepared and its morphological and mechanical properties were investigated by scanning electron microscopy (SEM) and material testing. The biocompatibility was evaluated by measuring the adhesion rate and cytotoxicity of bone marrow stromal cells (BMSCs). The strengthened ß-TCP/PRP composite had an appearance like the fungus Boletus kermesinus with the PRP gel distributed on the surface of the micropores. The maximum load and load intensity were 945.6±86.4 N and 13.1±0.5 MPa, which were significantly higher than those of ß-TCP (110.1±14.3 N and 1.6±0.2 MPa; P<0.05). The BMSC adhesion rate on the strengthened ß-TCP/PRP composite was >96% after 24 h, with a cell cytotoxicity value of zero. SEM micrographs revealed that following seeding of BMSCs onto the composite in high-glucose Dulbecco's modified Eagle's medium culture for two weeks, the cells grew well and exhibited fusiform, spherical and polygonal morphologies, as well as pseudopodial connections. The strengthened ß-TCP/PRP composite has the potential to be used as a scaffold in bone tissue engineering due to its effective biocompatibility and mechanical properties.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...