Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
J Biomed Nanotechnol ; 18(1): 61-76, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-35180900

RESUMEN

Kartogenin (KGN) can effectively promote the differentiation of adipose derived stem cells (ADSCs) into chondrocytes. With the help of three-dimensional slow-release technology, nano-microspheres are generated and used for cartilage repair. First, KGN solution was prepared, which was dissolved in distilled water, and NaOH solution, HEPES buffer, sodium chloride particles, and hydroxyapatite (HA) solution were added to prepare KGN-HA gel solution containing KGN. ADSCs were isolated from the posterior iliac of four-week-old New Zealand rabbits. After 0.5 mL of rabbit second-generation ADSCs suspension was taken, 2 mL KGN-HA gel solution was added, and they were mixed well to obtain ADSCs/KGN-HA gel. After drying treatment, ADSCs/KGN-HA nanospheres were precipitated. In the experiment, the minimum inhibitory concentration (MIC) of Staphylococcus aureus (MIC) > 2 µg/mL in each group of KGN-HA gel solution was reached within 30 days. Group K3 had the highest KGN encapsulation rate and the largest cumulative release. The biological activity of ADSCs was good in the ADSCs/KGN-HA nanoparticle solution. After two weeks of incubation, the nanospheres were positive for type II collagen staining/toluidine blue staining, that was, chondrocyte phenotype. The rabbit knee articular cartilage defect model was established. The defect part was filled with ADSCs/KGN-HA gel, which was similar in color to the surrounding tissues. The two sides of the tissue section and the surrounding cartilage tissue healed well, and no carrier material remained. Moreover, the cells were round, with cartilage lacuna formed around them, and after the simple periosteum was covered and repaired, the surface was sunken. The cell structure changed, and the healing with the surroundings was poor. In summary, under the slow release of KGN, ADSCs/KGN-HA nanospheres made ADSCs maintain a good biological form, which grew and proliferated normally. The ADSCs/KGN-HA nanoparticles cultured in vitro had a good repair effect on the animal model of articular cartilage defects.


Asunto(s)
Cartílago Articular , Durapatita , Anilidas , Animales , Cartílago Articular/cirugía , Células Cultivadas , Articulación de la Rodilla , Ácidos Ftálicos , Conejos , Células Madre , Ingeniería de Tejidos/métodos
4.
Drug Deliv ; 28(1): 319-324, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33517806

RESUMEN

The present work aims to examine the effect of gelatin on the stabilization of silver nanoparticles (AgNPs) and their use in healing the bone fracture. AgNPs-loaded Gel hydrogels (AgNPs/Gel) were fabricated under sunlight using gelatin (Gel) as stabilizing agent. The characterization of the synthesized hydrogels was performed with the help of techniques such as UV-visible spectroscopy (UV-Vis) and high-resolution transmission electron microscopy (HR-TEM). Furthermore, the results of cell cytotoxicity confirmed that the AgNPs/Gel hydrogels are nonhazardous to osteoblasts. The outcome of cell fixation with AgNPs/Gel hydrogels after an incubation period of five days exposed the improved survival and spreading of osteoblasts cells on the prepared AgNPs/Gel hydrogels. Moreover, the AgNPs/Gel hydrogel nanostructures displayed their ability in modulating bone fracture healing, which suggests their potential use in nursing care.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Fracturas Óseas/tratamiento farmacológico , Gelatina/química , Hidrogeles/química , Hidrogeles/farmacología , Nanopartículas del Metal/química , Plata/química , Animales , Células Cultivadas , Nanoestructuras/química , Osteoblastos/efectos de los fármacos , Porcinos , Cicatrización de Heridas/efectos de los fármacos
5.
Stem Cell Res Ther ; 11(1): 227, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32522250

RESUMEN

BACKGROUND: Bone marrow mesenchymal stem cells (BMSCs) exhibit the capacity to self-renew and differentiate into multi-lineage cell types, including osteoblasts, which are crucial regulators of fracture healing. Thus, this study aims to investigate the effect of microRNA (miR)-22-3p from BMSC-derived EVs on osteogenic differentiation and its underlying mechanism. METHODS: Extracellular vesicles (EVs) were isolated from BMSCs and taken up with BMSCs. Dual-luciferase reporter gene assay was used to verify the binding relationship between miR-22-3p and FTO. Loss- and gain-of-function experiments were performed to determine the roles of EV-delivered miR-22-3p and FTO in osteogenic differentiation as well as their regulatory role in the MYC/PI3K/AKT axis. To determine the osteogenic differentiation, ALP and ARS stainings were conducted, and the levels of RUNX2, OCN, and OPN level were determined. In vivo experiment was conducted to determine the function of EV-delivered miR-22-3p and FTO in osteogenic differentiation, followed by ALP and ARS staining. RESULTS: miR-22-3p expression was repressed, while FTO expression was elevated in the ovariectomized mouse model. Overexpression of miR-22-3p, EV-delivered miR-22-3p, increased ALP activity and matrix mineralization of BMSCs and promoted RUNX2, OCN, and OPN expressions in BMSCs. miR-22-3p negatively targeted FTO expression. FTO silencing rescued the suppressed osteogenic differentiation by EV-delivered miR-22-3p inhibitor. FTO repression inactivated the MYC/PI3K/AKT pathway, thereby enhancing osteogenic differentiation both in vivo and in vitro. CONCLUSION: In summary, miR-22-3p delivered by BMSC-derived EVs could result in the inhibition of the MYC/PI3K/AKT pathway, thereby promoting osteogenic differentiation via FTO repression.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , MicroARNs , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Diferenciación Celular , Células Cultivadas , Ratones , MicroARNs/genética , Osteogénesis/genética , Fosfatidilinositol 3-Quinasas
6.
J Cell Mol Med ; 24(9): 4944-4955, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32198976

RESUMEN

Bone defects resulting from non-union fractures or tumour resections are common clinical problems. Long non-coding RNAs (lncRNAs) are reported to play vital roles in stem cell differentiation. The aim of this study was to elucidate the role of lncRNA-H19 in osteogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs). Following the establishment of an osteogenic differentiation model in rats, the expression of H19, microRNA-149 (miR-149) and stromal cell-derived factor-1 (SDF-1) was measured by RT-qPCR. Thereafter, BMMSCs were isolated from rats and treated with a series of mimic, inhibitor or siRNA. SDF-1 expression, alkaline phosphatase (ALP) activity and osteocalcin (OCN) content were detected. The mineralized and calcified nodules were assessed by alizarin red S and Von Kossa staining. BMMSC surface markers were detected by flow cytometry. Western blot analysis was used to measure the expression of ALP, OCN, runt-related transcription factor 2 (RUNX2) and osterix (OSX) proteins. Lastly, dual-luciferase reporter gene assay and RNA immunoprecipitation were applied to verify the relationship of H19, miR-149 and SDF-1. Overexpressed H19 and SDF-1 and poorly expressed miR-149 were found in rats with osteogenic differentiation. H19 increased SDF-1 expression by binding to miR-149. H19 enhanced ALP activity, OCN content, calcium deposit and ALP, OCN, RUNX2 and OSX protein expression of BMMSCS by up-regulating SDF-1 via binding to miR-149. Taken together, up-regulated H19 could promote the osteogenic differentiation of BMMSCs by increasing SDF-1 via miR-149.


Asunto(s)
Células de la Médula Ósea/citología , Quimiocina CXCL12/metabolismo , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Regeneración Ósea , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Genes Reporteros , Masculino , Osteocalcina/biosíntesis , Osteogénesis , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/metabolismo , Transfección , Regulación hacia Arriba
7.
Clin Exp Rheumatol ; 38(6): 1088-1095, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32141427

RESUMEN

OBJECTIVES: Angiotensinogen (AGT) and miR-149-5p were differentially expressed genes in the osteoarthritis (OA), but their functional contribution to this disease is unclear. Our study aimed to illustrate their relevance to OA pathology and chondrocytic inflammation responses. METHODS: In this study, a total of 32 healthy donors and 56 OA patients were recruited for cartilage tissues, and interleukin (IL)-6-stimulated human chondrocyte-articular (HC-a) cells were used as an in vitro OA model. RESULTS: RT-qPCR and western blot assays demonstrated that AGT was upregulated in OA cartilage tissues while miR-149-5p was downregulated. Using a loss-of-function assay and inhibitor treatment, we found that AGT knockdown inhibited the increase of IL-1ß, matrix metalloproteinase (MMP)-13 and nitrite in IL-6-induced chondrocytes through blocking the renin-angiotensin system (RAS). The prediction (TargetScan) and validation (mutant and luciferase reporter assays) of the interaction between AGT and miR-149-5p indicated that miR-149-5p directly regulated inflammatory responses in OA chondrocytes by binding to AGT. Furthermore, using overexpression and inhibitor treatment experiments, our study proved that JAK2/STAT3 was activated in OA tissues, and AGT regulated OA inflammation via activating the JAK2/STAT3 pathway. CONCLUSIONS: Our study demonstrated that AGT, modulated and directly bond by miR-149-5p, promoted the IL-6-induced inflammatory responses in OA via JAK2/STAT3 pathway.


Asunto(s)
MicroARNs , Osteoartritis , Angiotensinógeno , Condrocitos , Genes , Humanos , Interleucina-1beta , Interleucina-6/genética , Janus Quinasa 2/genética , MicroARNs/genética , Osteoartritis/genética , Factor de Transcripción STAT3
8.
J Cell Physiol ; 235(4): 3634-3645, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31583718

RESUMEN

Exosomes derived from differentiated P12 cells and MSCs were proved to suppress apoptosis of neuron cells, and phosphatase and tensin homolog pseudogene 1 (PTENP1) was reported to inhibit cell proliferation. In this study, we aimed to investigate the role of PTENP1 in the process of post-spinal cord injury (SCI) recovery, so as to evaluate the therapeutic effects of exosomes derived from MSCs transfected with PTENP1 short hairpin RNA (shRNA), as a type of novel biomarkers in the treatment of SCI. Electron microscopy was used to observe the morphology of different exosomes. Real-time polymerase chain reaction and western blot, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, flow cytometry, Nissl staining, immunohistochemistry assay, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay were conducted to investigate and validate the underlying molecular signaling pathway. PTENP1-shRNA downregulated PTENP1 and PTEN while upregulating miR-21 and miR-19b. PTENP1-shRNA also accelerated cell apoptosis and reduced cell viability. In addition, PTENP1 reduced the miR-21 and miR-19b expression by directly targeting miR-21 and miR-19b. Meanwhile, both miR-21 and miR-19b reduced the expression of PTEN by directly targeting the 3'-untranslated region of PTEN. Furthermore, PTEN level and apoptosis index of neuron cells was the highest in the SCI group, while the treatment with exosomes+PTENP1-shRNA reduced the PTEN expression to a level similar to that in the sham group. Finally, PTENP1 inhibited miR-21 and miR-19b expression but upregulated PTEN expression. The upregulation of miR-21/miR-19b also suppressed the apoptosis of neuron cells by downregulating the PTEN expression. PTENP1 is involved in the recovery of SCI by regulating the expression of miR-19b and miR-21, and exosomes from PTENP1-shRNA-transfected cells may be used as a novel biomarker in SCI treatment.


Asunto(s)
MicroARNs/genética , Neuronas/metabolismo , ARN Largo no Codificante/genética , Traumatismos de la Médula Espinal/genética , Regiones no Traducidas 3'/genética , Apoptosis/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Exosomas/genética , Exosomas/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Neuronas/patología , Transducción de Señal , Traumatismos de la Médula Espinal/patología
9.
Front Cell Dev Biol ; 8: 581545, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33553139

RESUMEN

OBJECTIVE: Mesenchymal stem cells (MSCs) confer therapeutic benefits in various pathologies and cancers by releasing extracellular vesicles (EVs) loaded with bioactive compounds. Herein, we identified bone marrow MSC (BMSC)-derived EVs harboring microRNA (miR)-29b-3p to regulate osteogenic differentiation through effects on the suppressor of cytokine signaling 1 (SOCS1)/nuclear factor (NF)-κB pathway via targeting of lysine demethylase 5A (KDM5A) in osteoporosis. METHODS: We quantified the miR-29b-3p in BMSC-derived EVs from bone marrow specimens of osteoporotic patients and non-osteoporotic patients during total hip arthroplasty (THA). miR-29b-3p targeting KDM5A was confirmed by promoter luciferase assay, and enrichment of KDM5A in the promoter region of SOCS1 was analyzed by chromatin immunoprecipitation (ChIP). The expression and translocation of NF-κB to the nucleus were detected by western blot analysis and immunofluorescence staining, respectively. An ovariectomized (OVX) osteoporosis mouse model was established to further confirm the in vitro findings. RESULTS: BMSC-derived EVs of osteoporotic patients exhibited downregulated miR-29b-3p. EV-encapsulated miR-29b-3p from BMSCs potentiated osteogenic differentiation by specifically inhibiting KDM5A. KDM5A inhibited osteogenic differentiation by the regulation of H3K4me3 and H3K27ac of SOCS1. SOCS1 potentiated osteogenic differentiation by inhibiting NF-κB pathway. CONCLUSION: EV-encapsulated miR-29b-3p derived from BMSCs potentiated osteogenic differentiation through blockade of the SOCS1/NF-κB pathway by inhibition of KDM5A.

10.
Bioorg Med Chem Lett ; 29(20): 126665, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31495556

RESUMEN

Pyruvate dehydrogenase kinases (PDKs) act as negative modulator of mitochondrial pyruvate dehydrogenase complex (PDC) and play a crucial role in the regulation of oxidative glycolysis, which recently have been considered as a potential drug target for varying types of cancer and diabetes. Herein, we describe the discovery and biological validation of novel anti-osteosarcoma therapeutics targeting PDK2. We identified 14 anti-osteosarcoma compounds from an in-house small molecule library, which were then evaluated in a PDK2 kinase inhibition assay. We found that compounds with 2-((4-oxo-6-((4-phenylpiperazin-1-yl)methyl)-4H-pyran-3-yl)oxy)acetamide moiety showed promising inhibitory potencies to PDK2. Especial for 12, which bound to PDK2 with a Kd value of 2.3 µM, and inhibited PDK2 activity with an EC50 value of 1.1 µM. In addition, 12 selectively inhibited PDK2, the selectivity indexes are 10.6, 22.0, and 60.9 for PDK2 as compared to PDK1, 2 and 4, respectively. The MTT assay suggested that 12 reduced MG-63 cancer cell proliferation with an IC50 value of 4.7 µM. All these observations indicated that 12 was a novel anti-osteosarcoma therapeutic, which deserved for further investigation.


Asunto(s)
Acetamidas/química , Antineoplásicos/química , Osteosarcoma/tratamiento farmacológico , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/química , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Acetamidas/farmacología , Aminoácidos/química , Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Modelos Moleculares , Estructura Molecular , Terapia Molecular Dirigida , Piperazinas/química , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
12.
J Cell Physiol ; 234(12): 23485-23494, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31206187

RESUMEN

MicroRNAs (miRs) involve in osteogenic differentiation and osteogenic potential of mesenchymal stem cells (MSCs). Accordingly, the present study aimed to further uncover role miR-149 plays in osteogenic differentiation of MSCs with the involvement of the stromal cell-derived factor-1 (SDF-1)/CXC chemokine receptor-4 (CXCR4) pathway. Initially, the osteogenic differentiation model was induced. Next, the positive expression of STRO-1 in periosteum, alkaline phosphatase (ALP) activity, osteocalcin (OCN) protein content, and the calcium deposition in MSCs were determined. MSCs were treated with DNA methyltransferase inhibitor 5-aza-CdR, SDF-1 neutralizing antibody, or CXCR4 antagonist AMD3100 to investigate their roles in osteogenic differentiation; with the expression of CD44, CD90, CD14, and CD45 detected. Furthermore, the levels of SDF-1 and CXCR4, and the genes related to stemness (Nanog, Oct-4, and Sox-2) were measured to explore the effects of miR-149. The obtained data revealed the upregulation of STRO-1 in the periosteum. miR-149 could specifically bind to SDF-1. Besides, increased miR-149 methylation, higher ALP activity and OCN content, decreased positive rates of CD44 and CD90, and increased positive rates of CD14 and CD45 were found in osteogenic differentiation of MSCs. Subsequently, 5-Aza-CdR treatment reversed the above-mentioned effects. MSCs were finally treated with SDF-1 neutralizing antibody or AMD3100 to decrease Nanog, Oct-4, and Sox-2 expression. Taken together these results, miR-149 hypermethylation has the potential to activate the SDF-1/CXCR4 pathway and further promote osteogenic differentiation of MSCs.


Asunto(s)
Diferenciación Celular , Quimiocina CXCL12/metabolismo , Metilación de ADN , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Receptores CXCR4/metabolismo , Regiones no Traducidas 3' , Animales , Sitios de Unión , Células Cultivadas , Quimiocina CXCL12/genética , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/patología , MicroARNs/genética , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Osteoblastos/patología , Fenotipo , Ratas Sprague-Dawley , Receptores CXCR4/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal
13.
Oncol Lett ; 17(3): 2592-2598, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30854034

RESUMEN

Osteosarcoma (OS) is the most common bone malignancy, and is particularly prevalent in children and adolescents. OS is an aggressive tumor with a tendency to metastasize and invade to para-carcinoma tissues. The primary treatment for this tumor is a combination of surgery and chemotherapy. However, the prognosis remains poor due to chemoresistance and early metastasis. Osteopontin (OPN), a multifunctional secreted protein, has emerged as an important potential biomarker for diagnosing and treating cancer. The overexpression of OPN has been found in numerous malignant tumors, including breast, lung, gastric and ovarian cancer, as well as melanoma. Recent studies have suggested that OPN may provide an important function in the diagnosis and treatment of OS. The present review summarizes current knowledge and progress in understanding the potential role of OPN as a biomarker in OS.

14.
Asian Pac J Cancer Prev ; 17(8): 3785-91, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27644617

RESUMEN

BACKGROUND: Previous studies have assessed the association between the Cytotoxic T-lymphocyte Antigen- 4(CTLA-4) polymorphism with the risk of malignant bone tumor, but the conclusions were inconsistent. We aimed to clarify association of cytotoxic T-lymphocyte antigen-4 polymorphisms with malignant bone tumors risk by performing a meta-analysis. MATERIALS AND METHODS: The databases including PubMed, EMBase databases and the Cochrane Library were searched to identify the eligible studies prior to January 30 2016. Odds ratio (OR) with 95% confidence interval (95%CI) were used to estimate the strengths of the association between the CTLA-4 polymorphism and the malignant bone tumor risks. The meta-analysis was performed by STATA 12.0. RESULTS: Four individual studies with a total of 1003 cases with malignant bone tumors and 1162 controls were included in our meta-analysis. The results of meta-analysis on those data demonstrated that CTLA-4 +49G>A polymorphism was associated with the risk of Ewing's sarcoma and osteosarcoma strongly (A vs. G: : OR=1.36, 95%CI:1.20-1.54, p=0.000; AA+AG vs. GG: OR=1.35, 95%CI:1.14-1.61, p=0.001; AA vs. GG: OR=2.24, 95%CI:1.67-2.99, p=0.000; AA vs. AG+GG: OR=2.00, 95%CI:1.53-2.62, p=0.000), but CTLA-4 -318C/T polymorphism was not associated with the risk of malignant bone tumor (C vs. T: OR=0.76, 95%CI:0.76-1.08, p= 0.262; CC+CT vs. TT: OR=0.70, 95%CI:0.41-1.20, p=0.198; CC vs. TT: OR=0.69, 95%CI:0.40-1.19, p= 0.183; CC vs. CT+TT: OR=0.92, 95%CI:0.75-1.13, p= 0.419). Subgroup analysis showed that there are significantly positive correlations between CTLA-4 +49G>A polymorphism and increased risks of malignant bone tumors in large size of sample (A vs. G: OR=1.347, 95%CI: 1.172,1.548, p=0.000; AA vs. GG: OR=2.228, 95%CI: 1.608,3.085, p=0.000), Ewing's Sarcoma or Osteosarcoma (A vs. G: OR=1.361, 95%CI: 1.201,1.540, p=0.000; AA vs. GG: OR=2.236, 95%CI: 1.674,2.986, p=0.000), and PCR-RFLP or Sequencing(A vs. G: OR=1.361, 95%CI: 1.201,1.540, p=0.000; AA vs. GG: OR=2.236, 95%CI: 1.674,2.986, p=0.000), but CTLA-4 -318C/T polymorphism was not associated with the risk of malignant bone tumors in diagnosis, genotype method, and sample size (all p>0.05). CONCLUSIONS: CTLA-4 +49A/G variant was associated with an increased risk of developing the malignant bone tumors, such as Ewing's sarcoma and osteosarcoma. However, it failed to show any association between CTLA-4 -318C/T polymorphism and the risk of malignant bone tumors. Future large-scale studies remain to be done to confirm our conclusions.


Asunto(s)
Neoplasias Óseas/etiología , Neoplasias Óseas/genética , Antígeno CTLA-4/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Linfocitos T Citotóxicos/metabolismo , Adulto , Huesos/metabolismo , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Masculino , Osteosarcoma/etiología , Osteosarcoma/genética , Factores de Riesgo , Sarcoma de Ewing/etiología , Sarcoma de Ewing/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA