Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bioconjug Chem ; 27(2): 329-40, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26646666

RESUMEN

A bone imaging toolkit of 21 fluorescent probes with variable spectroscopic properties, bone mineral binding affinities, and antiprenylation activities has been created, including a novel linking strategy. The linking chemistry allows attachment of a diverse selection of dyes fluorescent in the visible to near-infrared range to any of the three clinically important heterocyclic bisphosphonate bone drugs (risedronate, zoledronate, and minodronate or their analogues). The resultant suite of conjugates offers multiple options to "mix and match" parent drug structure, fluorescence emission wavelength, relative bone affinity, and presence or absence of antiprenylation activity, for bone-related imaging applications.


Asunto(s)
Enfermedades Óseas/diagnóstico , Huesos/patología , Difosfonatos/química , Colorantes Fluorescentes/química , Imagen Óptica/métodos , Animales , Línea Celular , Humanos , Masculino , Ratas Sprague-Dawley
2.
Nat Genet ; 38(5): 525-7, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16642017

RESUMEN

Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disorder of skeletal malformations and progressive extraskeletal ossification. We mapped FOP to chromosome 2q23-24 by linkage analysis and identified an identical heterozygous mutation (617G --> A; R206H) in the glycine-serine (GS) activation domain of ACVR1, a BMP type I receptor, in all affected individuals examined. Protein modeling predicts destabilization of the GS domain, consistent with constitutive activation of ACVR1 as the underlying cause of the ectopic chondrogenesis, osteogenesis and joint fusions seen in FOP.


Asunto(s)
Receptores de Activinas Tipo I/genética , Mutación , Miositis Osificante/genética , Receptores de Activinas Tipo I/química , Secuencia de Aminoácidos , Animales , Cromosomas Humanos Par 2 , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , ARN Mensajero/genética , Homología de Secuencia de Aminoácido
3.
J Biol Chem ; 287(44): 36990-8, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22977237

RESUMEN

Bone morphogenetic protein (BMP) receptor kinases are tightly regulated to control development and tissue homeostasis. Mutant receptor kinase domains escape regulation leading to severely degenerative diseases and represent an important therapeutic target. Fibrodysplasia ossificans progressiva (FOP) is a rare but devastating disorder of extraskeletal bone formation. FOP-associated mutations in the BMP receptor ALK2 reduce binding of the inhibitor FKBP12 and promote leaky signaling in the absence of ligand. To establish structural mechanisms of receptor regulation and to address the effects of FOP mutation, we determined the crystal structure of the cytoplasmic domain of ALK2 in complex with the inhibitors FKBP12 and dorsomorphin. FOP mutations break critical interactions that stabilize the inactive state of the kinase, thereby facilitating structural rearrangements that diminish FKBP12 binding and promote the correct positioning of the glycine-serine-rich loop and αC helix for kinase activation. The balance of these effects accounts for the comparable activity of R206H and L196P. Kinase activation in the clinically benign mutant L196P is far weaker than R206H but yields equivalent signals due to the stronger interaction of FKBP12 with R206H. The presented ALK2 structure offers a valuable template for the further design of specific inhibitors of BMP signaling.


Asunto(s)
Receptores de Activinas Tipo I/química , Miositis Osificante/enzimología , Receptores de Activinas Tipo I/antagonistas & inhibidores , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Secuencias de Aminoácidos , Animales , Proteína Morfogenética Ósea 4/fisiología , Dominio Catalítico , Cristalografía por Rayos X , Activación Enzimática , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Ratones , Modelos Moleculares , Mutación Missense , Miositis Osificante/genética , Unión Proteica , Pirazoles/química , Pirimidinas/química , Transducción de Señal , Tacrolimus/farmacología , Proteína 1A de Unión a Tacrolimus/antagonistas & inhibidores , Proteína 1A de Unión a Tacrolimus/química , Proteína 1A de Unión a Tacrolimus/metabolismo
4.
J Mater Sci Mater Med ; 24(6): 1571-80, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23645077

RESUMEN

Porous scaffolds are limited in volume due to diffusion constraint and delay of vascular network formation. Channels have the potential to speed up cellular penetration. Their effectiveness in improving angiogenic cell penetration was assessed in vitro and in vivo in 3-D collagen scaffolds. In vitro, channelled and non-channelled scaffolds were seeded with vascular smooth muscle cells. Results demonstrated that the scaffolds supported angiogenic cell ingrowth in culture and the channels improved the depth of cell penetration into the scaffold (P < 0.05). The cells reside mainly around and migrate along the channels. In vivo, channels increased cell migration into the scaffolds (P < 0.05) particularly angiogenic cells (P < 0.05) resulting in a clear branched vascular network of microvessels after 2 weeks in the channelled samples which was not apparent in the non-channelled samples. Channels could aid production of tissue engineered constructs by offering the possibility of rapid blood vessel infiltration into collagen scaffolds.


Asunto(s)
Colágeno Tipo I/química , Microvasos/crecimiento & desarrollo , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Neovascularización Fisiológica/fisiología , Ingeniería de Tejidos/instrumentación , Andamios del Tejido , Animales , Proliferación Celular , Células Cultivadas , Colágeno Tipo I/ultraestructura , Análisis de Falla de Equipo , Femenino , Humanos , Ensayo de Materiales , Ratones , Ratones Endogámicos BALB C , Microvasos/citología , Diseño de Prótesis
5.
Cell Tissue Bank ; 14(2): 267-76, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22802140

RESUMEN

This study aimed to optimise methods of cryopreserving human articular cartilage (AC) tissue for the isolation of late chondrocytes. Human AC specimens from osteoarthritis patients who had undergone total knee replacement were used to optimise the chondrocyte isolation process and the choice of cryoprotective agent (CPA). For AC tissue cryopreservation, intact cored cartilage discs (5 mm diameter) and diced cartilage (0.2-1 mm cubes) from the same sized discs were step cooled and stored in liquid nitrogen for up to 48 h before chondrocyte isolation and in vitro assay of cell viability and proliferative potential. The results showed that 10 % dimethyl sulphoxide in 90 % foetal bovine serum was a successful CPA for chondrocyte cryopreservation. Compared with intact cored discs, dicing of AC tissue into 0.2-1 mm cubes significantly increased the viability and proliferative capacity of surviving chondrocytes after cryopreservation. In situ cross-section imaging using focused ion beam microscopy revealed that dicing of cored AC discs into small cubes reduced the cryo-damage to cartilage tissue matrix. In conclusion, modification of appropriate factors, such as the size of the tissue, cryoprotective agent, and isolation protocol, can allow successful isolation of viable chondrocytes with high proliferative capacity from cryopreserved human articular cartilage tissue. Further studies are required to determine whether these cells may retain cartilage differentiation capacity and provide sufficient chondrocytes for use as implants in clinical applications.


Asunto(s)
Cartílago Articular/patología , Proliferación Celular , Separación Celular/métodos , Condrocitos/patología , Criopreservación/métodos , Anciano , Anciano de 80 o más Años , Cartílago Articular/efectos de los fármacos , Cartílago Articular/fisiología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Crioprotectores/farmacología , Dimetilsulfóxido/farmacología , Femenino , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Osteoartritis de la Rodilla/patología , Osteoartritis de la Rodilla/fisiopatología , Temperatura
6.
Calcif Tissue Int ; 90(3): 202-10, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22249525

RESUMEN

Differences in the binding affinities of bisphosphonates for bone mineral have been proposed to determine their localizations and duration of action within bone. The main objective of this study was to test the hypothesis that mineral binding affinity affects bisphosphonate distribution at the basic multicellular unit (BMU) level within both cortical and cancellous bone. To accomplish this objective, skeletally mature female rabbits (n = 8) were injected simultaneously with both low- and high-affinity bisphosphonate analogs bound to different fluorophores. Skeletal distribution was assessed in the rib, tibia, and vertebra using confocal microscopy. The staining intensity ratio between osteocytes contained within the cement line of newly formed rib osteons or within the reversal line of hemiosteons in vertebral trabeculae compared to osteocytes outside the cement/reversal line was greater for the high-affinity compared to the low-affinity compound. This indicates that the low-affinity compound distributes more equally across the cement/reversal line compared to a high-affinity compound, which concentrates mostly near surfaces. These data, from an animal model that undergoes intracortical remodeling similar to humans, demonstrate that the affinity of bisphosphonates for the bone determines the reach of the drugs in both cortical and cancellous bone.


Asunto(s)
Conservadores de la Densidad Ósea/farmacocinética , Remodelación Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/metabolismo , Difosfonatos/farmacocinética , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Unión Competitiva/efectos de los fármacos , Unión Competitiva/fisiología , Remodelación Ósea/fisiología , Huesos/citología , Femenino , Osteón/citología , Osteón/efectos de los fármacos , Osteón/metabolismo , Osteocitos/citología , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Osteoporosis/tratamiento farmacológico , Conejos , Distribución Tisular/fisiología
7.
Eur J Cell Biol ; 87(6): 353-64, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18417247

RESUMEN

Human mesenchymal stem cells (hMSCs) from bone marrow were genetically marked by using a murine leukaemia virus construct encoding enhanced green fluorescent protein (eGFP). The marked cells were either directly implanted into the tibialis anterior muscle or introduced into a variety of other tissue sites in immunocompromised mice (NOD/SCID and C.B-17 SCID/beige) to investigate their fates and differentiation potentials. It was observed that the hMSCs survived for up to 12 weeks and showed site-specific morphological phenotypes. hMSCs delivered by intravenous injection were found mainly in the lungs and were detected rarely in other organs. Histomorphometry showed that, after implantation of hMSCs into the tibialis anterior muscle juxtaskeletally, the areas of reactive host callus formation at 1 and 2 weeks and of ectopic human bone formation at 1 week were significantly increased compared with the control group. Expression of eGFP and human RUNX2, alkaline phosphatase, osteocalcin, osteopontin, and collagen type I mRNAs were detected in mice implanted with the labelled hMSCs but not in sham-treated samples. Active clearance of the reactive callus and ectopic calcified tissue by osteoclast-like tartrate-resistant acid phosphatase-positive cells was observed. We conclude that the eGFP-labelled hMSCs can survive and retain the potential to differentiate morphologically into a variety of apparent mesenchymal phenotypes in vivo. Absolute confirmation of differentiation capacity requires further study and is complicated by known possibilities of fusion of donor and host cells or limited transfer of genetic material. Nevertheless, the genetically marked hMSCs are shown to participate extensively in bone formation and turnover. Control of the host osteoclast/macrophage responses resulting in clearance of formed osteogenic tissue warrants further investigation to promote prolonged human osteogenesis in immunocompromised mice. Furthermore, any proposed general cytotherapeutic strategy for enhanced osteogenesis is likely to require supplementation of local bone-forming biological signals.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Linaje de la Célula , Huésped Inmunocomprometido , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Osificación Heterotópica/metabolismo , Osteogénesis , Fosfatasa Alcalina/metabolismo , Animales , Células de la Médula Ósea/enzimología , Supervivencia Celular , Células Cultivadas , Colágeno Tipo I/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Huésped Inmunocomprometido/genética , Inmunohistoquímica , Células Madre Mesenquimatosas/enzimología , Ratones , Ratones SCID , Microscopía Fluorescente , Virus de la Leucemia Murina de Moloney/genética , Osificación Heterotópica/genética , Osificación Heterotópica/patología , Osteopontina/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Factores de Tiempo , Transfección
8.
Acta Biomater ; 4(5): 1322-31, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18440886

RESUMEN

Collagen-hydroxyapatite composites for bone tissue engineering are usually made by freezing an aqueous dispersion of these components and then freeze-drying. This method creates a foamed matrix which may not be optimum for growing cell colonies larger than a few hundred micrometres due to the limited diffusion of nutrients and oxygen, and the limited removal of waste metabolites. Incorporating a network of microchannels in the interior of the scaffold which may permit the flow of nutrient-rich media has been proposed as a method to overcome these diffusion constraints. A novel three-dimensional printing and critical point drying technique previously used to make collagen scaffolds has been modified to create collagen-hydroxyapatite scaffolds. This study investigates the properties of collagen and collagen-hydroxyapatite scaffolds and whether subjecting collagen and hydroxyapatite to critical point drying with liquid carbon dioxide results in any changes to the individual components. Specifically, the hydroxyapatite component was characterized before and after processing using wavelength-dispersive X-ray spectroscopy, X-ray diffraction and infrared spectroscopy. Critical point drying did not induce elemental, crystallographic or molecular changes in the hydroxyapatite. The quaternary structure of collagen was characterized using transmission electron microscopy and the quarter-staggered array characteristic of native collagen remained after processing. Microstructural characterization of the composites using scanning electron microscopy showed the hydroxyapatite particles were mechanically interlocked in the collagen matrix. The in vitro biological response of MG63 osteogenic cells to the composite scaffolds were characterized using the Alamar Blue, PicoGreen, alkaline phosphate and Live/Dead assays, and revealed that the critical point dried scaffolds were non-cytotoxic.


Asunto(s)
Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Dióxido de Carbono/química , Colágeno/química , Colágeno/farmacología , Durapatita/química , Durapatita/farmacología , Osteoblastos/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular , Desecación , Análisis de Falla de Equipo , Humanos , Osteoblastos/citología , Soluciones , Propiedades de Superficie
9.
J Med Chem ; 50(24): 5967-75, 2007 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-17975902

RESUMEN

Alpha-halogenated analogues of the anti-resorptive bisphosphonate risedronate (5, Ris) and its phosphonocarboxylate cognate (7, 3-PEHPC) were synthesized and compared with 5, 7, and the corresponding desoxy analogues in bone mineral affinity and mevalonate pathway inhibition assays. The Ris (5e-h) and 3-PEHPC (7e-h) analogues had decreased bone mineral affinity, confirming that the alpha-OH group in 5 and 7 enhances bone affinity. The 5 alpha-halo-analogues potently inhibited farnesyl pyrophosphate synthase (FPPS) with IC50 values from 16 (alpha-F) to 340 (alpha-Br) nM (5, 6 nM). In contrast, 7 alpha-halo-analogues were ineffective versus FPPS (IC50 > 600 microM), but inhibited Rab geranylgeranyl transferase (RGGT) (IC50 = 16-35 microM) similarly to 7 itself (IC50 = 24 microM). The alpha-F analogue 7e was 1-2 times as active as 7 in J774 cell viability and Rab11 prenylation inhibition assays.


Asunto(s)
Conservadores de la Densidad Ósea/síntesis química , Difosfonatos/síntesis química , Ácido Etidrónico/análogos & derivados , Organofosfonatos/síntesis química , Propionatos/síntesis química , Piridinas/síntesis química , Transferasas Alquil y Aril/antagonistas & inhibidores , Animales , Conservadores de la Densidad Ósea/química , Conservadores de la Densidad Ósea/farmacología , Línea Celular , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Cromatografía Liquida , Difosfonatos/química , Difosfonatos/farmacología , Durapatita/química , Ácido Etidrónico/síntesis química , Ácido Etidrónico/química , Ácido Etidrónico/farmacología , Geraniltranstransferasa/antagonistas & inhibidores , Ratones , Organofosfonatos/química , Organofosfonatos/farmacología , Propionatos/química , Propionatos/farmacología , Prenilación de Proteína/efectos de los fármacos , Piridinas/química , Piridinas/farmacología , Ácido Risedrónico , Relación Estructura-Actividad
10.
Ann N Y Acad Sci ; 1117: 209-57, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18056045

RESUMEN

The bisphosphonates (BPs) are well established as the treatments of choice for disorders of excessive bone resorption, including Paget's disease of bone, myeloma and bone metastases, and osteoporosis. There is considerable new knowledge about how BPs work. Their classical pharmacological effects appear to result from two key properties: their affinity for bone mineral and their inhibitory effects on osteoclasts. Mineral binding affinities differ among the clinically used BPs and may influence their differential distribution within bone, their biological potency, and their duration of action. The inhibitory effects of the nitrogen-containing BPs (including alendronate, risedronate, ibandronate, and zoledronate) on osteoclasts appear to result from their inhibition of farnesyl pyrophosphate synthase (FPPS), a key branch-point enzyme in the mevalonate pathway. FPPS generates isoprenoid lipids used for the posttranslational modification of small GTP-binding proteins essential for osteoclast function. Effects on other cellular pathways, such as preventing apoptosis in osteocytes, are emerging as other potentially important mechanisms of action. As a class, BPs share several common properties. However, as with other classes of drugs, there are obvious chemical, biochemical, and pharmacological differences among the various individual BPs. Each BP has a unique profile that may help to explain potential important clinical differences among the BPs, in terms of speed of onset of fracture reduction, antifracture efficacy at different skeletal sites, and the degree and duration of suppression of bone turnover. As we approach the 40th anniversary of the discovery of their biological effects, there remain further opportunities for using their properties for medical purposes.


Asunto(s)
Difosfonatos/química , Difosfonatos/farmacología , Osteoclastos/metabolismo , Animales , Neoplasias Óseas/secundario , Resorción Ósea , Huesos/metabolismo , Difosfonatos/uso terapéutico , Guanosina Trifosfato/química , Humanos , Modelos Biológicos , Modelos Químicos , Mieloma Múltiple/metabolismo , Metástasis de la Neoplasia , Nitrógeno/química , Osteocitos/metabolismo , Osteoporosis/terapia , Procesamiento Proteico-Postraduccional , Linfocitos T/metabolismo , Resultado del Tratamiento
11.
Biomater Transl ; 3(1): 1-2, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35837347
12.
Biotechnol Adv ; 35(4): 407-418, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28450077

RESUMEN

The plastic-adherent, fibroblast-like, clonogenic cells found in the human body now defined as multipotent "Mesenchymal Stromal Cells" (MSCs) hold immense potential for cell-based therapies. Recently, research and basic knowledge of these cells has fast-tracked, both from fundamental and translational perspectives. There have been important discoveries with respect to the available variety of tissue sources, the development of protocols for their easy isolation and in vitro expansion and for directed differentiation into various cell types. In addition, there has been discovery of novel abilities such as immune-modulation and further development of the use of biomaterials to aid isolation, expansion and differentiation together with improved delivery to the selected optimal tissue site. However, the molecular fingerprint of MSCs in these contexts remains imprecise and inadequate. Consequently, without this crucial knowledge it is difficult to achieve progress to determine with precision their practical developmental potentials. Detailed investigations on the global gene expression, or transcriptome, of MSCs could offer essential clues in this regard. In this article, we address the challenges associated with MSC transcriptome studies, the paradoxes observed in published experimental results and the need for careful transcriptomic analysis. We describe the exemplary applications with various transcriptome platforms that are used to address the variation in biomarkers and the identification of differentiation processes. The evolution and the potentials for adapting next-generation sequencing (NGS) technology in transcriptome analysis are discussed. Lastly, based on review of the existing understanding and published studies, we propose how NGS may be applied to promote further understanding of the biology of MSCs and their use in allied fields such as regenerative medicine.


Asunto(s)
Perfilación de la Expresión Génica , Células Madre Mesenquimatosas , Transcriptoma , Diferenciación Celular , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/tendencias , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Medicina Regenerativa
13.
J Bone Miner Res ; 32(9): 1860-1869, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28337806

RESUMEN

Bisphosphonates are widely used in the treatment of clinical disorders characterized by increased bone resorption, including osteoporosis, Paget's disease, and the skeletal complications of malignancy. The antiresorptive potency of the nitrogen-containing bisphosphonates on bone in vivo is now recognized to depend upon two key properties, namely mineral binding affinity and inhibitory activity on farnesyl pyrophosphate synthase (FPPS), and these properties vary independently of each other in individual bisphosphonates. The better understanding of structure activity relationships among the bisphosphonates has enabled us to design a series of novel bisphosphonates with a range of mineral binding properties and antiresorptive potencies. Among these is a highly potent bisphosphonate, 1-fluoro-2-(imidazo-[1,2 alpha]pyridin-3-yl)-ethyl-bisphosphonate, also known as OX14, which is a strong inhibitor of FPPS, but has lower binding affinity for bone mineral than most of the commonly studied bisphosphonates. The aim of this work was to characterize OX14 pharmacologically in relation to several of the bisphosphonates currently used clinically. When OX14 was compared to zoledronate (ZOL), risedronate (RIS), and minodronate (MIN), it was as potent at inhibiting FPPS in vitro but had significantly lower binding affinity to hydroxyapatite (HAP) columns than ALN, ZOL, RIS, and MIN. When injected i.v. into growing Sprague Dawley rats, OX14 was excreted into the urine to a greater extent than the other bisphosphonates, indicating reduced short-term skeletal uptake and retention. In studies in both Sprague Dawley rats and C57BL/6J mice, OX14 inhibited bone resorption, with an antiresorptive potency equivalent to or greater than the comparator bisphosphonates. In the JJN3-NSG murine model of myeloma-induced bone disease, OX14 significantly prevented the formation of osteolytic lesions (p < 0.05). In summary, OX14 is a new, highly potent bisphosphonate with lower bone binding affinity than other clinically relevant bisphosphonates. This renders OX14 an interesting potential candidate for further development for its potential skeletal and nonskeletal benefits. © 2017 American Society for Bone and Mineral Research.


Asunto(s)
Difosfonatos/farmacología , Difosfonatos/farmacocinética , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley
14.
Biomaterials ; 27(26): 4557-65, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16720039

RESUMEN

Depending upon local conditions, brushite (CaHPO4 x 2 H2O) cements may be largely resorbed or (following hydrolysis to hydroxyapatite) remain stable in vivo. To determine which factors influence cement resorption, previous studies have investigated the solution-driven degradation of brushite cements in vitro in the absence of any cells. However, the mechanism of cell-mediated biodegradation of the brushite cement is still unknown. The aim of the current study was to observe the cell-mediated biodegradation of brushite cement formulations in vitro. The cements were aged in the presence of a murine cell line (RAW264.7), which had the potential to form osteoclasts in the presence of the receptor for nuclear factor kappa B ligand (RANKL) in vitro, independently of macrophage colony stimulating factor (M-CSF). The cytotoxicity of the cements on RAW264.7 cells and the calcium and phosphate released from materials to the culture media were analysed. Scanning electron microscopy (SEM) and focused ion beam (FIB) microscopy were used to characterise the ultrastructure of the cells. The results showed that the RAW264.7 cell line formed multinucleated TRAP positive osteoclast-like cells, capable of ruffled border formation and lacunar resorption on the brushite calcium phosphate cement in vitro. In the osteoclast-like cell cultures, ultrastructural analysis by SEM revealed phenotypic characteristics of osteoclasts including formation of a sealing zone and ruffled border. Penetration of the surface of the cement, was demonstrated using FIB, and this showed the potential demineralising effect of the cells on the cements. This study has set up a useful model to investigate the cell-mediated cement degradation in vitro.


Asunto(s)
Materiales Biocompatibles/metabolismo , Cementos para Huesos/metabolismo , Fosfatos de Calcio/metabolismo , Macrófagos/metabolismo , Animales , Materiales Biocompatibles/toxicidad , Biodegradación Ambiental , Cementos para Huesos/toxicidad , Fosfatos de Calcio/toxicidad , Línea Celular , Ratones , Microscopía , Microscopía Electrónica de Rastreo , Modelos Biológicos
15.
Tissue Eng ; 12(9): 2521-31, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16995785

RESUMEN

The creation of a vascularized bed makes the survival of seeded cells on 3-dimensional scaffolds much more likely. However, relying purely on random capillary ingrowth into the porous scaffolds from the host may compromise vascularization of a scaffold. One solution is to transplant cells capable of differentiating into new blood vessels into the scaffolds to accelerate the creation of a vascularized scaffold. Because endothelial cells are the key cells involved in blood vessel formation, the present study was designed to investigate the development of a biomaterial surface that supports endothelial cell attachment and proliferation. The subsequent effects of the material surface modifications on the differentiation and proliferation of human bone marrow-derived fibroblasts (HBMFs) when grown in co-culture with a human bone marrow endothelial cell line (HBMEC-60) were studied. Endothelialization studies showed that the gelatin-coated and hydroxyapatite-coated substrates were superior for HBMEC-60 attachment and proliferation to hydrolyzed-only or untreated polycaprolactone substrates. Co-culture studies showed that the presence of the HBMEC-60 specifically enhanced HBMF cell proliferation and differentiation and that this effect was not observed with co-culture with skin fibroblasts. It is concluded that the co-culture of endothelial cells with HBMFs could be a promising culture system for bone tissue- engineering applications.


Asunto(s)
Sustitutos de Huesos , Diferenciación Celular/fisiología , Células Endoteliales/fisiología , Fibroblastos/fisiología , Neovascularización Fisiológica , Poliésteres , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Adhesión Celular/fisiología , Línea Celular , Materiales Biocompatibles Revestidos , Técnicas de Cocultivo , Células Endoteliales/citología , Fibroblastos/citología , Humanos , Especificidad de Órganos , Piel/citología , Piel/metabolismo , Ingeniería de Tejidos/métodos
16.
J Biomed Mater Res A ; 79(3): 582-90, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16817218

RESUMEN

Biodegradation of poly-DL-lactide-co-glycolide (PLGA) both in vitro and in vivo has been well documented. However, the roles that macrophages and their fused multinucleated giant cells (MNGCs) play in this biodegradation are still unclear. The current study aimed to investigate macrophage-mediated biodegradation of PLGA thin films and of PLGA composites with hydroxyapatite (HA) and tricalcium phosphate (TCP) ceramic powders in vitro using a murine macrophage cell line (RAW 264.7). The interactions were analyzed by using cell viability assays, scanning electron microscopy, and focused ion beam microscopy. The results showed that RAW 264.7 cells effectively attached and proliferated on the PLGA films and PLGA-HA, PLGA-TCP composites. The RAW 264.7 cells were observed to aggregate and fuse to form MNGCs. The cell processes on the membrane, or pseudopodia, penetrated into the PLGA films and evidently eroded the surface. We conclude that macrophages and fused MNGCs actively respond to PLGA films as substratum and degrade the surface of this polymer.


Asunto(s)
Ácido Láctico/metabolismo , Macrófagos/metabolismo , Ácido Poliglicólico/metabolismo , Polímeros/metabolismo , Animales , Fosfatos de Calcio/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ácido Láctico/toxicidad , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Microscopía Electrónica de Rastreo , Ácido Poliglicólico/toxicidad , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polímeros/toxicidad , Solventes
17.
Biomater Transl ; 2(2): 89-90, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35836964
19.
Biomater Transl ; 2(4): 285-286, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35837418
20.
Biomaterials ; 26(29): 5790-800, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15882901

RESUMEN

Human mesenchymal stem cells (hMSCs) were transfected using four retroviral pseudotypes, amphotropic murine leukemia viruses 4070 (MuLV-10A1), a modification of amphotropic pseudotype 4073 (A71G, Q74K, V139M), gibbon ape leukemia virus (GaLV), or feline endogenous virus (RD114) encoding the neomycin resistance (Neo(r)) gene and enhanced green fluorescent protein (eGFP) as genetic markers. It was observed that the MuLV4073 was the most efficient pseudotype for hMSC transfection. The proliferation and differentiation characteristics of eGFP-labelled hMSCs were not significantly different from control hMSCs. G418 selected eGFP-labelled cells were cultured for 3 weeks on two porous, commercially available calcium phosphate bioceramics, a "synthetic hydroxyapatite" and a "deproteinised bone", before implantation into NOD/SCID mice for up to 4 weeks. The eGFP-labelled hMSCs could be readily visualised by their intense green fluorescence both in vitro and in vivo. In "synthetic hydroxyapatite" implants the cells remained in a monolayer, whereas in "deproteinised bone" implants mineralised tissues were detected by histology, scanning electron microscopy and energy dispersive X-ray spectrometry. From the results, it is concluded that the use of eGFP-labelled hMSCs is an effective tool to trace the fate of hMSCs and evaluate the interactions between cells and ceramics both in vitro and in vivo. This is of great value in prospective assessments of these cell populations for use in tissue engineering applications.


Asunto(s)
Materiales Biocompatibles/química , Cerámica/química , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Mesenquimatosas/citología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Microscopía Electrónica de Rastreo , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrofotometría , Factores de Tiempo , Ingeniería de Tejidos , Transfección , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA