Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 457
Filtrar
1.
Histochem Cell Biol ; 155(3): 369-380, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33175185

RESUMEN

In this study, we examined the immunolocalization of podoplanin/E11, CD44, actin filaments, and phosphorylated ezrin in the osteoblasts on the verge of differentiating into osteocytes in murine femora and tibiae. When observing under stimulated emission depletion microscopy, unlike podoplanin-negative osteoblasts, podoplanin-positive osteoblasts showed a rearranged assembly of actin filaments along the cell membranes which resembled that of embedded osteocytes. In the metaphysis, i.e., the bone remodeling site, CD44-bearing osteoclasts were either proximal to or in contact with podoplanin-positive osteoblasts, but the podoplanin-positive osteoblasts also localized CD44 on their own cell surface. These podoplanin-positive osteoblasts, which either possessed CD44 on their cell surface or were close to CD44-bearing osteoclasts, showed phosphorylated ezrin-positivity on the cell membranes. Therefore, the CD44/podoplanin interaction on the cell surface may be involved in the osteoblastic differentiation into osteocytes in the metaphyses, via the mediation of podoplanin-driven ezrin phosphorylation and the subsequent reorganized assembly of actin filaments. Consistently, the protein expression of phosphorylated ezrin was increased after CD44 administration in calvarial culture. Conversely, in modeling sites such as the cortical bones, podoplanin-positive osteoblasts were uniformly localized at certain intervals even without contact with CD44-positive bone marrow cells; furthermore, they also exhibited phosphorylated ezrin immunoreactivity along their cell membranes. Taken together, it seems likely that the CD44/podoplanin interaction is involved in osteoblastic differentiation into osteocytes in the bone remodeling area but not in modeling sites.


Asunto(s)
Huesos/citología , Glicoproteínas de Membrana/análisis , Osteoblastos/citología , Osteocitos/citología , Animales , Remodelación Ósea , Huesos/química , Diferenciación Celular , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos ICR , Osteoblastos/química , Osteocitos/química
2.
Mol Med Rep ; 22(4): 3549-3558, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32945514

RESUMEN

Our previous study revealed that treatment with a combination of fibroblast growth factor­2 and melatonin (MEL) synergistically augmented osteogenic activity and mineralization of MC3T3­E1 mouse preosteoblast cells. Thus, the objective of the present study was to assess the effect of MEL on osteogenetic characteristics in human osteoblastic cells. Human jawbone­derived osteoblastic (hOB) cells were isolated from mandibular bone fragments. RUNX family transcription factor 2 (Runx2) expression, alkaline phosphatase (ALP) enzyme activity and the mineralization ability of hOB cells in the presence of MEL were evaluated. Microarray analysis was also performed to assess the expression of MEL­induced microRNAs (miRNAs/miRs) in hOB cells. Treatment with MEL significantly enhanced Runx2 expression, ALP activity and mineralization staining. However, this effect was significantly reduced following transforming growth factor­ß1 treatment. In total, 124 miRNAs were differentially expressed in MEL­treated hOB cells, compared with untreated cells. Of the upregulated miRNAs, miR­181c­5p exhibited the largest fold change. Runx2 mRNA expression and mineralization staining in the presence of MEL were significantly reduced following transfection with a miR­181c­5p inhibitor. In addition, transfection with miR-181c-5p mimics significantly increased Runx2 expression and mineralization staining. These results suggested that MEL­induced miR­181c­5p was involved in osteogenic differentiation and mineralization of hOB cells. Using TargetScan, a putative miR­181c­5p binding site was identified in the Notch2 gene. Moreover, Notch2 mRNA and protein expression levels in hOB cells were significantly reduced following transfection with miR­181c­5p mimics, confirming Notch2 as a target gene for miR­181c­5p. Notch2 siRNA knockdown significantly increased Runx2 expression and mineralization staining, which suggested that Notch2 may negatively regulate osteogenic differentiation of hOB cells by downregulating Runx2. In conclusion, MEL­induced expression of miR­181c­5p enhanced osteogenic differentiation and calcification of hOB cells.


Asunto(s)
Maxilares/citología , Melatonina/farmacología , MicroARNs/genética , Osteogénesis , Fosfatasa Alcalina/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Maxilares/química , Maxilares/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/química , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Adulto Joven
3.
J Cell Mol Med ; 24(18): 10792-10802, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32803867

RESUMEN

Brain-derived neurotrophic factor (BDNF) has been reported to participate in fracture healing, whereas the mechanism is still unclear. Since osteoblast migration is important for fracture healing, investigating effects of BDNF on osteoblasts migration may help to reveal its mechanism. Here, MC3T3-E1 cells were used in vitro while closed femur fracture mice were applied in vivo. Cells migration was assessed with Transwell assay. The protein expression was analysed by immunoblotting. X-ray and Micro-CT were performed at different time after fracture. Our results showed that BDNF promoted MC3T3-E1 cells migration, integrin ß1 expression and ERK1/2 and AKT phosphorylation. K252a, a specific inhibitor for TrkB, suppressed BDNF-induced migration, integrin ß1 expression and activation of ERK1/2 and AKT. PD98059 (an ERK1/2 inhibitor) and LY294002 (an AKT inhibitor) both inhibited BDNF-induced migration and integrin ß1 expression while integrin ß1 blocking antibody only suppressed cell migration. X-ray and Micro-CT analyses showed that the adenoviral carried integrin ß1 shRNA group had slower fracture healing at 7 and 21 days, but not 35 days compared to the control group. Thus, we proposed that BDNF stimulated MC3T3-E1 cells migration by up-regulating integrin ß1 via TrkB mediated ERK1/2 and AKT signalling, and this may help to enhance the fracture healing.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Curación de Fractura/efectos de los fármacos , Integrina beta1/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Glicoproteínas de Membrana/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Línea Celular Transformada , Movimiento Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/análisis , Fracturas del Fémur/diagnóstico por imagen , Fracturas del Fémur/tratamiento farmacológico , Fracturas del Fémur/fisiopatología , Integrina beta1/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/química , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Regulación hacia Arriba/efectos de los fármacos , Microtomografía por Rayos X
4.
Artículo en Inglés | MEDLINE | ID: mdl-32769062

RESUMEN

Asp, Glu, and D-Ser are chiral amino acids and neurotransmitters binding to the N-methyl-D-aspartate receptor (NMDA) and they participate in glutamate signalization. D-amino acids are increasingly being recognized as important signaling molecules and variations in their levels are considered a marker of different pathologies, however, there is still a lack of knowledge about the role of most of D-amino acids in living organisms such as bone cells. A method for determination of concentrations of L/D-Asp, L/D-Glu and L/D-Ser in two types of bone cell lines: murine osteocytes (MLOY4) and osteoblasts (MC3T3-E1) is presented. It is based on capillary electrophoresis coupled to laser-induced fluorescence detection in normal polarity with 4-fluoro-7-nitro-2,1,3-benzoxadiazole as derivatizing agent suitable for an Argon ion laser source. The electrolyte consists of 137.5 mM borate buffer and 12.5 mM ß-cyclodextrins as chiral selectors and the separation lasts 25 min. The method was optimized and validated for specificity, sensitivity, linearity, accuracy, and precision in murine osteocytes and osteoblasts. LLOQ was 0.25 µmol L-1 for the three D-amino acids and linearity was confirmed with r > 0.995 for all D-and L-amino acids. Accuracy ranged between 81.9% and 111.7% and intra-day precision ranged between 1.8% and 10.9%. Concentrations of D- and L- Asp, Glu, and Ser are given and statistical differences between osteocytes and osteoblasts were found. The highest differences corresponded to L- and D-Glu. This method could play a fundamental role in the study of therapeutic targets in the treatment of bone diseases.


Asunto(s)
Aminoácidos/análisis , Electroforesis Capilar/métodos , Osteoblastos/química , Osteocitos/química , Animales , Línea Celular , Límite de Detección , Modelos Lineales , Ratones , Reproducibilidad de los Resultados , Espectrometría de Fluorescencia
5.
Handb Exp Pharmacol ; 262: 231-258, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32661663

RESUMEN

Chemokines are a family of small proteins, subdivided by their conserved cysteine residues and common structural features. Chemokines interact with their cognate G-protein-coupled receptors to elicit downstream signals that result in cell migration, proliferation, and survival. This review presents evidence for how the various CXC and CC subfamily chemokines influence bone hemostasis by acting on osteoclasts, osteoblasts, and progenitor cells. Also discussed are the ways in which chemokines contribute to bone loss as a result of inflammatory diseases such as rheumatoid arthritis, HIV infection, and periodontal infection. Both positive and negative effects of chemokines on bone formation and bone loss are presented. In addition, the role of chemokines in altering the bone microenvironment through effects on angiogenesis and tumor invasion is discussed. Very few therapeutic agents that influence bone formation by targeting chemokines or chemokine receptors are available, although a few are currently being evaluated.


Asunto(s)
Quimiocinas/química , Infecciones por VIH , Huesos , Quimiocinas/inmunología , Humanos , Osteoblastos/química , Osteoblastos/fisiología , Osteoclastos/química , Osteoclastos/fisiología
6.
J Biomater Appl ; 35(3): 385-404, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32567484

RESUMEN

One of the most common prophylactic techniques to solve prosthetic joint infection (PJI) is incorporation of antibiotics into acrylic bone cement to prevent bacterial colonization and proliferation by providing local antibiotic delivery directly at the implant site. Further, there has been a significant concern over the efficacy of commonly used antibiotics within bone cement due to the rise in multi-drug resistant (MDR) microorganisms. Selenium is an essential trace element that has multiple beneficial effects for human health and its chemotherapeutic action is well known. It was reported that nanostructured selenium enhanced bone cell adhesion and has an increased osteoblast function. In this context, we used the selenium nanoparticles (SeNPs) to improve antibacterial and antioxidant properties of poly (methyl methacrylate) (PMMA) and tri calcium phosphate (TCP)-based bone cements, and to reduce of the infection risk caused by orthopedic implants. As another novelty of this study, we proposed phosphatidylcholine (PC) as a unique and natural stabilizer in the synthesis of selenium nanoparticles. After the structural analysis of the prepared bone cements was performed, in vitro osteointegration and antibacterial efficiency were tested using MC3T-E1 (mouse osteoblastic cell line) and SaOS-2 (human primary osteogenic sarcoma) cell lines, and S. aureus (Gram positive) and E.coli (Gram negative) strains, respectively. More importantly, PC-SeNPs-reinforced bone cements exhibited significant effect against E. coli, compared to S. aureus and a dose-dependent antibacterial activity against both bacterial strains tested. Meanwhile, these bone cements induced the apoptosis of SaOS-2 through increased reactive oxygen species without negatively influencing the viability of the healthy cell line. Furthermore, the obtained confocal images revealed that PC-SeNPs (103.7 ± 0.56 nm) altered the cytoskeletal structure of SaOS-2 owing to SeNPs-induced apoptosis, when MC3T3-E1 cells showed a typical spindle-shaped morphology. Taken together, these results highlighted the potential of PC-SeNPs-doped bone cements as an effective graft material in bone applications.


Asunto(s)
Antibacterianos/química , Cementos para Huesos/química , Nanopartículas/química , Fosfatidilcolinas/química , Selenio/química , Animales , Antibacterianos/farmacología , Antioxidantes/química , Apoptosis/efectos de los fármacos , Fosfatos de Calcio/química , Línea Celular , Escherichia coli/efectos de los fármacos , Humanos , Ratones , Osteoblastos/química , Osteoblastos/metabolismo , Polimetil Metacrilato/química , Especies Reactivas de Oxígeno/química , Selenio/farmacología , Staphylococcus aureus/efectos de los fármacos
7.
Mol Med Rep ; 22(3): 1737-1746, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32582985

RESUMEN

Circular RNAs (circRNAs) are a class of non­coding RNAs that exhibit important regulatory roles in various biological processes. However, the role of circRNAs and their potential role in osteoblast differentiation and mineralization is unclear. The aim of the present study was to investigate the expression of mmu_circ_003795 and its effect on collagen type XV α 1 chain (COL15A1). First, it was identified that the expression levels of mmu_circ_003795 and osteopontin (OPN) were upregulated in the induced cells. Silencing of mmu_circ_003795 reduced the gene and protein levels of COL15A1 and OPN, whereas the expression level of mmu­microRNA (miR)­1249­5p was upregulated. In addition, after 7 or 14 days of induction, alkaline phosphatase and Alizarin Red­S staining were decreased in the mmu_circRNA_003795 inhibitory group compared with the negative control group. In conclusion, mmu_circ_003795 may regulate osteoblast differentiation and mineralization in MC3T3­E1 and MDPC23 cells via mmu­miR­1249­5p by targeting COL15A1.


Asunto(s)
Colágeno/genética , Colágeno/metabolismo , Osteoblastos/citología , ARN Circular/genética , Células 3T3 , Animales , Diferenciación Celular , Línea Celular , Regulación de la Expresión Génica , Ratones , MicroARNs/genética , Osteoblastos/química , Osteogénesis , Osteopontina/genética , Regulación hacia Arriba
8.
Biotechniques ; 68(6): 311-317, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32301333

RESUMEN

Extracting sufficient quantity and quality RNA from bone is essential for downstream application, such as transcriptomic sequencing, to evaluate gene expression. Isolation of RNA from bone presents a unique challenge owing to the hypocellular, brittle and mineralized matrix, which makes homogenizing the tissue difficult and provides little RNA to work with. Removal of contaminating tissue, such as bone marrow and connective tissue, is essential for isolating RNA that is unique to osteoblasts, osteoclasts and osteocytes. This study established a method to effectively isolate RNA from normal canine bone cells using the phalanges, without contamination from other tissue types, for downstream transcriptomic analysis.


Asunto(s)
Huesos/química , Biología Molecular/métodos , ARN/aislamiento & purificación , Transcriptoma/genética , Animales , Perros , Regulación de la Expresión Génica/genética , Osteoblastos/química , Osteoclastos/química , Osteocitos/química , ARN/química , ARN/genética
9.
J Colloid Interface Sci ; 572: 408-420, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32272315

RESUMEN

BACKGROUND AND AIM: Membranes for guided bone regeneration should have a mechanical structure and a chemical composition suitable for mimicking biological structures. In this work, we pursue the development of periosteum-inspired bilayered membranes obtained by crosslinking alginate with different amounts of nanohydroxyapatite. EXPERIMENTS: Alginate-nanohydroxyapatite interaction was studied by rheology and infrared spectroscopy measurements. The membranes were characterized regarding their tensile strength, degradation and surface morphology. Finally, cell cultures were performed on each side of the membranes. FINDINGS: The ionic bonding between alginate polysaccharide networks and nanohydroxyapatite was proven, and had a clear effect in the strength and microstructure of the hydrogels. Distinct surface characteristics were achieved on each side of the membranes, resulting in a highly porous fibrous side and a mineral-rich side with higher roughness and lower porosity. Moreover, the effect of amount of nanohydroxyapatite was reflected in a decrease of the membranes' plasticity and an increment of degradation rate. Finally, it was proved that osteoblast-like cells proliferated and differentiated on the mineral-rich side, specially when a higher amount of nanohydroxyapatite was used, whereas fibroblasts-like cells were able to proliferate on the fibrous side. These periosteum-inspired membranes are promising biomaterials for guided tissue regeneration applications.


Asunto(s)
Alginatos/química , Materiales Biomiméticos/química , Durapatita/química , Membrana Dobles de Lípidos/química , Nanopartículas/química , Periostio/química , Diferenciación Celular , Células Cultivadas , Humanos , Osteoblastos/química , Osteoblastos/citología , Tamaño de la Partícula , Propiedades de Superficie
10.
Handb Exp Pharmacol ; 262: 177-230, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32006259

RESUMEN

Cytokines and hematopoietic growth factors have traditionally been thought of as regulators of the development and function of immune and blood cells. However, an ever-expanding number of these factors have been discovered to have major effects on bone cells and the development of the skeleton in health and disease (Table 1). In addition, several cytokines have been directly linked to the development of osteoporosis in both animal models and in patients. In order to understand the mechanisms regulating bone cells and how this may be dysregulated in disease states, it is necessary to appreciate the diverse effects that cytokines and inflammation have on osteoblasts, osteoclasts, and bone mass. This chapter provides a broad overview of this topic with extensive references so that, if desired, readers can access specific references to delve into individual topics in greater detail.


Asunto(s)
Citocinas , Osteoporosis , Animales , Huesos , Humanos , Osteoblastos/química , Osteoclastos/química
11.
Int J Mol Sci ; 21(1)2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-31906530

RESUMEN

Fused deposit modeling (FDM) 3D printing technology cannot generate scaffolds with high porosity while maintaining good integrity, anatomical-surface detail, or high surface area-to-volume ratio (S/V). Solvent casting and particulate leaching (SCPL) technique generates scaffolds with high porosity and high S/V. However, it is challenging to generate complex-shaped scaffolds; and solvent, particle and residual water removal are time consuming. Here we report techniques surmounting these problems, successfully generating a highly porous scaffold with the anatomical-shape characteristics of a human femur by polylactic acid polymer (PLA) and PLA-hydroxyapatite (HA) casting and salt leaching. The mold is water soluble and is easily removable. By perfusing with ethanol, water, and dry air sequentially, the solvent, salt, and residual water were removed 20 fold faster than utilizing conventional methods. The porosities are uniform throughout the femoral shaped scaffold generated with PLA or PLA-HA. Both scaffolds demonstrated good biocompatibility with the pre-osteoblasts (MC3T3-E1) fully attaching to the scaffold within 8 h. The cells demonstrated high viability and proliferation throughout the entire time course. The HA-incorporated scaffolds demonstrated significantly higher compressive strength, modulus and osteoinductivity as evidenced by higher levels of alkaline-phosphatase activity and calcium deposition. When 3D printing a 3D model at 95% porosity or above, our technology preserves integrity and surface detail when compared with FDM-generated scaffolds. Our technology can also generate scaffolds with a 31 fold larger S/V than FDM. We have developed a technology that is a versatile tool in creating personalized, patient-specific bone graft scaffolds efficiently with high porosity, good scaffold integrity, high anatomical-shaped surface detail and large S/V.


Asunto(s)
Materiales Biocompatibles/química , Osteoblastos/química , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Materiales Biocompatibles/síntesis química , Calcio/análisis , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Fuerza Compresiva , Durapatita/química , Fémur , Humanos , Ensayo de Materiales , Osteoblastos/enzimología , Osteoblastos/metabolismo , Perfusión , Poliésteres/química , Porosidad , Andamios del Tejido/efectos adversos
12.
Int J Mol Sci ; 21(3)2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31979313

RESUMEN

Titanium (Ti) is an osteoconductive material that is routinely used as a bulk implant to fix and restore bones and teeth. This study explored the effective use of Ti as a bone engineering scaffold. Challenges to overcome were: (1) difficult liquid/cell infiltration into Ti microfiber scaffolds due to the hydrophobic nature of Ti; and (2) difficult cell attachment on thin and curved Ti microfibers. A recent discovery of UV-photofunctionalization of Ti prompted us to examine its effect on Ti microfiber scaffolds. Scaffolds in disk form were made by weaving grade 4 pure Ti microfibers (125 µm diameter) and half of them were acid-etched to roughen the surface. Some of the scaffolds with original or acid-etched surfaces were further treated by UV light before cell culture. Ti microfiber scaffolds, regardless of the surface type, were hydrophobic and did not allow glycerol/water liquid to infiltrate, whereas, after UV treatment, the scaffolds became hydrophilic and immediately absorbed the liquid. Osteogenic cells from two different origins, derived from the femoral and mandibular bone marrow of rats, were cultured on the scaffolds. The number of cells attached to scaffolds during the early stage of culture within 24 h was 3-10 times greater when the scaffolds were treated with UV. The development of cytoplasmic projections and cytoskeletal, as well as the expression of focal adhesion protein, were exclusively observed on UV-treated scaffolds. Osteoblastic functional phenotypes, such as alkaline phosphatase activity and calcium mineralization, were 2-15 times greater on UV-treated scaffolds, with more pronounced enhancement on acid-etched scaffolds compared to that on the original scaffolds. These effects of UV treatment were associated with a significant reduction in atomic carbon on the Ti microfiber surfaces. In conclusion, UV treatment of Ti microfiber scaffolds tunes their physicochemical properties and effectively enhances the attachment and function of osteoblasts, proposing a new strategy for bone engineering.


Asunto(s)
Oseointegración , Osteoblastos/metabolismo , Andamios del Tejido/química , Titanio/efectos de la radiación , Animales , Células de la Médula Ósea/citología , Calcificación Fisiológica/fisiología , Técnicas de Cultivo de Célula , Células Cultivadas , Fémur/citología , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Mandíbula/citología , Microscopía Electrónica de Rastreo , Osteoblastos/química , Osteoblastos/enzimología , Osteogénesis/fisiología , Ratas , Ratas Sprague-Dawley , Propiedades de Superficie/efectos de la radiación , Ingeniería de Tejidos , Titanio/química , Rayos Ultravioleta
13.
Analyst ; 145(3): 764-776, 2020 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-31755889

RESUMEN

Bone mineral development has been described to proceed through an amorphous precursor prior to apatite crystallization. However, further analytical approaches are necessary to identify specific markers of amorphous mineral components in bone. Here, we establish an original Fourier transform infrared (FTIR) spectroscopy approach to allow the specific identification of the amorphous and/or crystalline nature of bone mineral. Using a series of standards, our results demonstrate that obtaining the second derivative of the FTIR spectra could reveal a peak specifically corresponding to amorphous calcium phosphate (ACP) at ∼992 cm-1. The intensity of this peak was strongly correlated to ACP content in standard mixtures. The analysis of a variety of bones showed that a clear ACP peak could be identified as a specific marker of the existence of an amorphous mineral component in developing bones. In contrast, the ACP peak was not detected in the mature bones. Moreover, subjecting developing bones to ex vivo crystallization conditions led to a clear reduction of the ACP peak, further substantiating the conversion of amorphous mineral precursor into mature apatite crystals. Analysis of mineralization in osteogenic cell cultures corroborated our observations, showing the presence of ACP as a major transient component in early mineralization, but not in the mature matrix. Additionally, FTIR imaging revealed that ACP was present in areas of matrix development, distributed around the edges of mineralizing nodules. Using an original analytical approach, this work provides strong evidence to support that bone mineral development is initiated by an amorphous precursor prior to apatite crystallization.


Asunto(s)
Huesos/química , Fosfatos de Calcio/química , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Animales , Huesos/metabolismo , Línea Celular , Ratones , Ratones Endogámicos C57BL , Osteoblastos/química , Osteoblastos/citología , Osteoblastos/metabolismo , Ratas , Ratas Sprague-Dawley , Pez Cebra
14.
Stem Cell Res Ther ; 10(1): 402, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852542

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) are multipotent stem cells that are able to differentiate into several cell types, including cartilage, fat, and bone. As a common progenitor, MSC differentiation has to be tightly regulated to maintain the balance of their differentiation commitment. It has been reported that the decision process of MSCs into fat and bone cells is competing and reciprocal. Several factors have been suggested as critical factors that affect adipo-osteogenic decision, including melatonin and smad4. Yes-associated protein (YAP) is an important effector protein in the Hippo signaling pathway that acts as a transcriptional regulator by activating the transcription of the genes involved in cell proliferation and anti-apoptosis. The non-canonical role of YAP in regulating bone homeostasis by promoting osteogenesis and suppressing adipogenesis was recently demonstrated in a mouse model. However, it is unclear whether YAP is also crucial for modulating human MSC differentiation to fat and bone. METHODS: The expression level of YAP during MSC differentiation was modulated using pharmaceutical molecule and genetic experiments through gain- and loss-of-function approaches. RESULTS: We demonstrated for the first time that YAP has a non-canonical role in regulating the balance of adipo-osteogenic differentiation of human MSCs. The result from synchrotron radiation-based Fourier transform infrared (FTIR) microspectroscopy showed unique metabolic fingerprints generated from YAP-targeted differentiated cells that were clearly distinguished from non-manipulated control. CONCLUSIONS: These results, thus, identify YAP as an important effector protein that regulates human MSC differentiation to fat and bone and suggests the use of FTIR microspectroscopy as a promising technique in stem cell research.


Asunto(s)
Adipogénesis , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Osteogénesis , Factores de Transcripción/metabolismo , Adipocitos/química , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dobutamina/farmacología , Humanos , Inmunofenotipificación , Lisofosfolípidos/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/química , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Análisis de Componente Principal , Espectroscopía Infrarroja por Transformada de Fourier , Factores de Transcripción/genética , Cordón Umbilical/citología
15.
Sci Rep ; 9(1): 16130, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31695092

RESUMEN

Abnormal antioxidative capabilities were observed in the pathogenesis of steroid-induced osteoporosis (SIOP). Ferroptosis is a recently discovered type of cell death that is characterized by the overproduction of ROS in response to GPX4 and system Xc- downregulation, which is mediated by an Fe2+ fenton reaction. However, investigations focusing on the relationship between ferroptosis and steroid-induced bone disease remain limited. In the present study, high-dose dexamethasone was used to establish a mouse SIOP model, and extracellular vesicles extracted from bone marrow-derived endothelial progenitor cells (EPC-EVs) alleviated the pathological changes in SIOP via microtomography (micro-CT), with elevations in bone volume (BV), bone surface (BS), trabecular thickness (Tb.Th), and trabecular connectivity density (Conn-D) and decreases in trabecular separation (Tb.sp) and the structure model index (SMI). Histopathological analysis, such as haematoxylin and eosin (HE) and Masson staining, showed that EPC-EVs treatment increased the volume and density of the trabecular bone and bone marrow. RNA sequencing (RNA-seq) and bioinformatics analysis revealed subcellular biological alterations upon steroid and EPC-EVs treatment. Compared with the control, high-dose dexamethasone downregulated GPX4 and system XC-, and the Kyoto Encyclopedia of Genes and Genomes (KEGG)-based gene set enrichment analysis suggested that the ferroptotic pathway was activated. In contrast, combination treatment with EPC-EVs partly reversed the KEGG-mapped changes in the ferroptotic pathway at both the gene and mRNA expression levels. In addition, alterations in ferroptotic marker expression, such as SLC3A2, SLC7A11, and GPX4, were further confirmed by RNA-seq. EPC-EVs were able to reverse dexamethasone treatment-induced alterations in cysteine and several oxidative injury markers, such as malondialdehyde (MDA), glutathione (GSH), and glutathione disulphide (GSSG) (as detected by ELISA). In conclusion, EPC-EVs prevented mouse glucocorticoid-induced osteoporosis by suppressing the ferroptotic pathway in osteoblasts, which may provide a basis for novel therapies for SIOP in humans.


Asunto(s)
Dexametasona/efectos adversos , Células Progenitoras Endoteliales/química , Vesículas Extracelulares/química , Ferroptosis , Osteoporosis/prevención & control , Animales , Densidad Ósea , Células Progenitoras Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Humanos , Malondialdehído/metabolismo , Ratones , Osteoblastos/química , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoporosis/etiología , Osteoporosis/metabolismo , Osteoporosis/fisiopatología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo
16.
Biofabrication ; 12(1): 015007, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31509811

RESUMEN

To successfully achieve the porous cell-blocks, a bioink is a prerequisite requirement. However, although various hydrogel-based bioinks have been applied, a hydrogel/bioceramic-based composite bioink consisting of cells has not been actively investigated owing to its poor printability and low initial cell-viability. In this study, a new bioink consisting of fibrillated collagen, cells, and bioceramic (ß-TCP) is suggested to attain a 3D porous cell-laden composite structure with high cellular responses, in aspects of initial cell viability, proliferation, and differentiation using preosteoblasts (MC3T3-E1) and human adipose stem cells (hASCs). By manipulating the processing conditions and weight fractions of the ceramic in the bioink, a 3D porous cell-laden composite structure can be fabricated successfully. The cell-laden composite structure revealed that the printed structure was mechanically stable, the laden cells were satisfactorily viable, and even cell proliferation/differentiation was well performed. Moreover, the cells in the composite structure exhibited significant osteogenic activities compared to the pure collagen bioink (control), and higher levels of osteogenic gene expression of the hASC-laden composite structure were observed without using an osteogenic medium than those of the control using an osteogenic medium, indicating that the laden ß-TCP triggered osteogenic differentiation of the hASCs.


Asunto(s)
Bioimpresión/métodos , Huesos/citología , Colágeno/química , Ingeniería de Tejidos/métodos , Adipocitos/química , Adipocitos/citología , Adipocitos/metabolismo , Animales , Bioimpresión/instrumentación , Regeneración Ósea , Huesos/metabolismo , Proliferación Celular , Supervivencia Celular , Cerámica/química , Colágeno/metabolismo , Humanos , Hidrogeles/química , Ratones , Osteoblastos/química , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Porosidad , Impresión Tridimensional , Células Madre/citología , Ingeniería de Tejidos/instrumentación , Andamios del Tejido/química
17.
Biofabrication ; 12(1): 015005, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31443097

RESUMEN

Cells with differentiation potential into mesodermal types are the focus of emerging bone tissue engineering (TE) strategies as an alternative autologous source. When the source of cells is extremely limited or not readily accessible, such as in severe injuries, a tissue biopsy may not yield the required number of viable cells. In line, adipose-derived stromal cells (ASCs) quickly became attractive for bone TE, since they can be easily and repeatably harvested using minimally invasive techniques with low morbidity. Inspired by the multiphenotypic cellular environment of bone, we propose the co-encapsulation of ASCs and osteoblasts (OBs) in self-regulated liquefied and multilayered microcapsules. We explore the unique architecture of such hybrid units to provide a dynamic environment using a simple culture in spinner flasks. Results show that microtissues were successfully obtained inside the proposed microcapsules with an appropriate diffusion of essential molecules for cell survival and signaling. Remarkably, microcapsules cultured in the absence of supplemental osteogenic differentiation factors presented osteopontin immunofluorescence, evidencing that the combined effect of the dynamic environment, and the paracrine signaling between ASCs and OBs may prompt the development of bone-like microtissues. Furthermore, microcapsules cultured under dynamic environment presented an enhanced mineralized matrix and a more organized extracellular matrix ultrastructure compared to static cultures used as control. Altogether, data in this study unveil an effective engineered bioencapsulation strategy for the in vitro production of bone-like microtissues in a more realistic and cost-effective manner. Accordingly, we intend to use the proposed system as hybrid devices implantable by minimally invasive procedures for bone TE applications.


Asunto(s)
Adipocitos/citología , Huesos/citología , Osteoblastos/citología , Ingeniería de Tejidos/métodos , Adipocitos/química , Adipocitos/metabolismo , Huesos/química , Huesos/metabolismo , Diferenciación Celular , Matriz Extracelular/química , Humanos , Osteoblastos/química , Osteoblastos/metabolismo , Osteopontina/metabolismo , Células del Estroma/química , Células del Estroma/citología , Células del Estroma/metabolismo , Ingeniería de Tejidos/instrumentación , Andamios del Tejido/química
18.
Anal Chem ; 91(16): 10395-10400, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31318197

RESUMEN

Protein and peptide adhesion is a major factor contributing to sample loss during proteomic sample preparation workflows. Sample loss often has detrimental effects on the quality of proteomic analysis by compromising protein identification and data reproducibility. When starting with a low sample amount, only the most abundant proteins can be identified, which often offers little insights for biological research. Although the general idea about severe sample loss from low amount of starting material is widely presumed in the proteomics field, quantitative assessment on the impact of sample loss has been poorly investigated. In the present study, we have quantitatively assessed sample loss during each step of a conventional in-solution sample preparation workflow using bicinchoninic acid (BCA) and targeted LC/MS/MS protein and peptide assays. According to our assessment, for starting materials of ∼1000 mammalian cells, surface adhesion, along with desalting and speed-vacuum drying steps, all contribute heavily to sample loss, in particular for low-abundance proteins. With this knowledge, we have adapted slippery liquid infused porous surface (SLIPS) treatment, commercial LoBind tubes, and in-line desalting during sample processing. With these improvements, we were able to use a conventional in-solution sample handling method to identify on average 829 proteins with 1000 U2OS osteosarcoma cells (∼100 ng) with 75-min LC/MS/MS runs, an 11-fold increase in protein identification. Our optimized in-solution workflow is straightforward and also much less equipment- and technique-demanding than other advanced sample preparation protocols in the field.


Asunto(s)
Osteoblastos/química , Péptidos/aislamiento & purificación , Proteínas/aislamiento & purificación , Proteómica/métodos , Línea Celular Tumoral , Cromatografía Liquida , Humanos , Quinolinas/química , Reproducibilidad de los Resultados , Manejo de Especímenes/normas , Espectrometría de Masas en Tándem , Flujo de Trabajo
19.
J Histochem Cytochem ; 67(10): 723-734, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31238004

RESUMEN

Primary cilia, hair-like organelles projecting from the surface of cells, are critical for sensing extracellular stimuli and transmitting molecular signals that regulate cell functions. During bone development, cell cilia are found in several types of cells, but their roles require further investigation. Intraflagellar transport (IFT) is essential for the formation and maintenance of most eukaryotic cilia. IFT140 is a core protein of the IFT-A complex. Mutations in IFT140 have been associated with cases of skeletal ciliopathies. In this study, we examined the expression of IFT140 during bone development. The results showed that, compared with many soft tissues, Ift140 (mRNA level) was highly expressed in bone. Moreover, its expression level was downregulated in the long bones of murine osteoporosis models. At the histological level, IFT140 was characteristically expressed in osteoblasts and chondrocytes at representative stages of bone development, and its expression level in these two types of cells was observed in two waves. These findings suggest that IFT140 may play an important role in the process of chondrogenic and osteogenic differentiation during bone development.


Asunto(s)
Desarrollo Óseo/genética , Huesos/metabolismo , Proteínas Portadoras/genética , Expresión Génica , Animales , Huesos/química , Huesos/embriología , Proteínas Portadoras/análisis , Proteínas Portadoras/fisiología , Diferenciación Celular , Células Cultivadas , Condrocitos/química , Condrocitos/ultraestructura , Cilios/química , Regulación hacia Abajo , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Osteoblastos/química , Osteoblastos/ultraestructura , Osteogénesis/genética , Osteoporosis/genética , ARN Mensajero/análisis
20.
Eur Rev Med Pharmacol Sci ; 23(11): 4521-4529, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31210279

RESUMEN

OBJECTIVE: We aimed at detecting the expression of long non-coding ribonucleic acid (lncRNA) maternally expressed 3 (MEG3) in the serum of fracture patients, and at investigating its impacts on the proliferation and differentiation of osteoblasts and the specific molecular mechanism of action. PATIENTS AND METHODS: The serum samples of 48 fracture patients diagnosed in our hospital (Fracture group) and 30 healthy people receiving physical examination (Health group) were collected. The expression level of serum lncRNA MEG3 in Fracture group and Health group was measured via reverse transcription-polymerase chain reaction (RT-PCR). Furthermore, a small interfering RNA (siRNA) was applied to construct mouse osteoblast cell line MC3T3-E1 with a stable knockout of MEG3. The growth status of the cell was observed, and the impacts of MEG3 knockout on the osteoblast proliferation were determined using cell counting kit-8 (CCK-8), a proliferation activity detection kit. Meanwhile, 5-ethynyl-2'-deoxyuridine (EdU) staining was applied to detect the proportion of EdU positive cells in the osteoblasts in Control group and MEG3 knockout group (MEG3 siRNA group). In addition, RT-PCR was performed to measure the messenger RNA (mRNA) levels of differentiation-related genes. Finally, RT-PCR and Western blotting assay were adopted to analyze the expression of the Wnt/ß-catenin signaling pathway. RESULTS: The expression of serum lncRNA MEG3 in fracture patients was increased markedly (p<0.05). Results of in-vitro cell experiment indicated that intervention with MEG3 siRNA could obviously promote the proliferation and differentiation of osteoblast cell line MC3T3-E1. The results of RT-PCR and Western blotting assay revealed that the role of MEG3 in promoting differentiation and proliferation might be mediated by the activation of the Wnt/ß-catenin signaling pathway in osteoblasts. CONCLUSIONS: LncRNA MEG3 can promote the proliferation and differentiation of osteoblasts by activating the Wnt/ß-catenin signaling pathway, so it is expected to become a new target for accelerating the fracture healing.


Asunto(s)
Fracturas Óseas/genética , Osteoblastos/citología , ARN Largo no Codificante/genética , Adulto , Animales , Estudios de Casos y Controles , Diferenciación Celular , Línea Celular , Proliferación Celular , Fracturas Óseas/sangre , Técnicas de Inactivación de Genes , Humanos , Ratones , Persona de Mediana Edad , Osteoblastos/química , ARN Largo no Codificante/sangre , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA