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
1.
J Craniomaxillofac Surg ; 47(2): 341-348, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30579746

RESUMEN

BACKGROUND: Cranioplasty is a surgical procedure used to treat a bone defect or deformity in the skull. To date, there is little consensus on the standard-of-care for graft materials used in such a procedure. Graft materials must have sufficient mechanical strength to protect the underlying brain as well as the ability to integrate and support new bone growth. Also, the ideal graft material should be individually customized to the contours of the defect to ensure a suitable aesthetic outcome for the patient. PURPOSE: Customized 3D-printed scaffolds comprising of polycaprolactone-ß-tricalcium phosphate (PCL-TCP) have been developed with mechanical properties suitable for cranioplasty. Osteostimulation of PCL-TCP was enhanced through the addition of a bone matrix-mimicking heparan sulphate glycosaminoglycan (HS3) with increased affinity for bone morphogenetic protein-2 (BMP-2). Efficacy of this PCL-TCP/HS3 combination device was assessed in a rat critical-sized calvarial defect model. METHOD: Critical-sized defects (5 mm) were created in both parietal bones of 19 Sprague Dawley rats (Male, 450-550 g). Each cranial defect was randomly assigned to 1 of 4 treatment groups: (1) A control group consisting of PCL-TCP/Fibrin alone (n = 5); (2) PCL-TCP/Fibrin-HSft (30 µg) (n = 6) (HSft is the flow-through during HS3 isolation that has reduced affinity for BMP-2); (3) PCL-TCP/Fibrin-HS3 (5 µg) (n = 6); (4) PCL-TCP/Fibrin-HS3 (30 µg) (n = 6). Scaffold integration and bone formation was evaluated 12-weeks post implantation by µCT and histology. RESULTS: Treatment with PCL-TCP/Fibrin alone (control) resulted in 23.7% ± 1.55% (BV/TV) of the calvarial defect being filled with new bone, a result similar to treatment with PCL-TCP/Fibrin scaffolds containing either HSft or HS3 (5 µg). At increased amounts of HS3 (30 µg), enhanced bone formation was evident (BV/TV = 38.6% ± 9.38%), a result 1.6-fold higher than control. Further assessment by 2D µCT and histology confirmed the presence of enhanced bone formation and scaffold integration with surrounding host bone only when scaffolds contained sufficient bone matrix-mimicking HS3. CONCLUSION: Enhancing the biomimicry of devices using a heparan sulphate with increased affinity to BMP-2 can serve to improve the performance of PCL-TCP scaffolds and provides a suitable treatment for cranioplasty.


Asunto(s)
Materiales Biomiméticos/uso terapéutico , Fosfatos de Calcio/uso terapéutico , Heparitina Sulfato/uso terapéutico , Poliésteres/uso terapéutico , Cráneo/cirugía , Andamios del Tejido , Animales , Materiales Biomiméticos/administración & dosificación , Fosfatos de Calcio/administración & dosificación , Heparitina Sulfato/administración & dosificación , Humanos , Imagenología Tridimensional , Masculino , Poliésteres/administración & dosificación , Ratas , Ratas Sprague-Dawley , Cráneo/diagnóstico por imagen
2.
Biomaterials ; 184: 41-55, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30205243

RESUMEN

Bone morphogenetic proteins (BMPs) are essential during tissue repair and remodeling after injury. Glycosaminoglycan (GAG) sugars are known to enhance BMP activity in vitro and in vivo; here the interactions of BMP-2 with various glycosaminoglycan classes were compared and shown to be selective for heparin over other comparable saccharides. The minimal chain lengths and specific sulfate moieties required for heparin-derived oligosaccharide binding to BMP-2, and the ability of such oligosaccharides to promote BMP-2-induced osteogenic differentiation in vitro were then determined. BMP-2 could bind to heparin hexasaccharides (dp6) and octasaccharides (dp8), but decasaccharides (dp10) were the minimum chain length required for both efficient binding of BMP-2 and consequent heparin-dependent cell responses. N-sulfation is the most important, and 6-O-sulfation moderately important for BMP-2 binding and activity, whereas 2-O-sulfation was much less critical. Bone formation assays in vivo further confirmed that dp10, N-sulfated heparin oligosaccharides were the minimal requirement for effective enhancement of BMP-2-induced bone formation. Such information is necessary for the rational design of the next generations of heparan-based devices for bone tissue repair.


Asunto(s)
Proteína Morfogenética Ósea 2/química , Heparina/química , Animales , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular , Línea Celular , Femenino , Heparitina Sulfato/química , Ratones , Osteogénesis , Unión Proteica , Estabilidad Proteica , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Propiedades de Superficie
3.
J Biomed Mater Res B Appl Biomater ; 106(5): 1887-1896, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28941021

RESUMEN

Functionalizing medical devices with polypeptides to enhance their performance has become important for improved clinical success. The extracellular matrix (ECM) adhesion protein vitronectin (VN) is an effective coating, although the chemistry used to attach VN often reduces its bioactivity. In vivo, VN binds the ECM in a sequence-dependent manner with heparan sulfate (HS) glycosaminoglycans. We reasoned therefore that sequence-based affinity chromatography could be used to isolate a VN-binding HS fraction (HS9) for use as a coating material to capture VN onto implant surfaces. Binding avidity and specificity of HS9 were confirmed by enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR)-based assays. Plasma polymerization of allylamine (AA) to tissue culture-treated polystyrene (TCPS) was then used to capture and present HS9 as determined by radiolabeling and ELISA. HS9-coated TCPS avidly bound VN, and this layered surface supported the robust attachment, expansion, and maintenance of human pluripotent stem cells. Compositional analysis demonstrated that 6-O- and N-sulfation, as well as lengths greater than three disaccharide units (dp6) are critical for VN binding to HS-coated surfaces. Importantly, HS9 coating reduced the threshold concentration of VN required to create an optimally bioactive surface for pluripotent stem cells. We conclude that affinity-purified heparan sugars are able to coat materials to efficiently bind adhesive factors for biomedical applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1887-1896, 2018.


Asunto(s)
Materiales Biocompatibles Revestidos/química , Proteínas de la Matriz Extracelular/química , Heparitina Sulfato/química , Células Madre Pluripotentes/metabolismo , Vitronectina/química , Adhesión Celular , Línea Celular , Humanos , Células Madre Pluripotentes/citología
4.
J Cell Physiol ; 232(3): 566-575, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27291835

RESUMEN

The future of human mesenchymal stem cells (hMSCs) as a successful cell therapy relies on bioprocessing strategies to improve the scalability of these cells without compromising their therapeutic ability. The culture-expansion of hMSCs can be enhanced by supplementation with growth factors, particularly fibroblast growth factor 2 (FGF2). The biological activity of FGF2 is controlled through interactions with heparan sulfate (HS) that facilitates ligand-receptor complex formation. We previously reported on an FGF2-interacting HS variant (termed HS2) isolated from embryonic tissue by anionic exchange chromatography that increased the proliferation and potency of hMSCs. Here, we detail the isolation of an FGF2 affinity-purified HS variant (HS8) using a scalable platform technology previously employed to generate HS variants with increased affinity for BMP-2 or VEGF165 . This process used a peptide sequence derived from the heparin-binding domain of FGF2 as a substrate to affinity-isolate HS8 from a commercially available source of porcine mucosal HS. Our data show that HS8 binds to FGF2 with higher affinity than to FGF1, FGF7, BMP2, PDGF-BB, or VEGF165 . Also, HS8 protects FGF2 from thermal destabilization and increases FGF signaling and hMSC proliferation through FGF receptor 1. Long-term supplementation of cultures with HS8 increased both hMSC numbers and their colony-forming efficiency without adversely affecting the expression of hMSC-related cell surface antigens. This strategy further exemplifies the utility of affinity-purifying HS variants against particular ligands important to the stem cell microenvironment and advocates for their addition as adjuvants for the culture-expansion of hMSCs destined for cellular therapy. J. Cell. Physiol. 232: 566-575, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Secuencia de Aminoácidos , Anticoagulantes/farmacología , Proliferación Celular , Cromatografía de Afinidad , Disacáridos/análisis , Factor Xa/metabolismo , Factor 2 de Crecimiento de Fibroblastos/química , Heparitina Sulfato/aislamiento & purificación , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Péptidos/química , Péptidos/metabolismo , Estabilidad Proteica/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Biomaterials ; 35(25): 6776-86, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24854095

RESUMEN

The therapeutic use of VEGF165 to stimulate blood vessel formation for the treatment of peripheral arterial disease or cardiovascular-related disease has met with limited success. Here we describe an affinity-isolated heparan sulfate glycotherapeutic (HS7(+ve)) that binds to, and enhances the bioactivity of, VEGF165. Application of HS7(+ve) complexed with VEGF165 results in enhanced VEGF165-VEGFR2 interaction, prolonged downstream pErk1/2 signalling, and increased cell proliferation and tube formation in HUVECs, compared with VEGF165 alone. The pro-angiogenic potential of HS7(+ve) was further assessed in vivo using the chick embryo chorioallantoic membrane (CAM) assay. Exogenous dosing with HS7(+ve) alone significantly enhanced the formation of new blood vessels with potencies comparable to VEGF165. These results demonstrate the potential for vascular therapy of glycotherapeutic agents targeted at augmenting the bioactivity of VEGF165.


Asunto(s)
Heparitina Sulfato/farmacología , Ingeniería de Proteínas/métodos , Enfermedades Vasculares/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/farmacología , Técnicas Biosensibles , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/química , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Unión Proteica , Transducción de Señal
6.
Bone ; 50(4): 954-64, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22227436

RESUMEN

Lowering the efficacious dose of bone morphogenetic protein-2 (BMP-2) for the repair of critical-sized bone defects is highly desirable, as supra-physiological amounts of BMP-2 have an increased risk of side effects and a greater economic burden for the healthcare system. To address this need, we explored the use of heparan sulfate (HS), a structural analog of heparin, to enhance BMP-2 activity. We demonstrate that HS isolated from a bone marrow stromal cell line (HS-5) and heparin each enhances BMP-2-induced osteogenesis in C2C12 myoblasts through increased ALP activity and osteocalcin mRNA expression. Commercially available HS variants from porcine kidney and bovine lung do not generate effects as great as HS5. Heparin and HS5 influence BMP-2 activity by (i) prolonging BMP-2 half-life, (ii) reducing interactions between BMP-2 with its antagonist noggin, and (iii) modulating BMP2 distribution on the cell surface. Importantly, long-term supplementation of HS5 but not heparin greatly enhances BMP-2-induced bone formation in vitro and in vivo. These results show that bone marrow-derived HS effectively supports bone formation, and suggest its applicability in bone repair by selectively facilitating the delivery and bioavailability of BMP-2.


Asunto(s)
Células de la Médula Ósea/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Heparitina Sulfato/farmacología , Osteogénesis/efectos de los fármacos , Animales , Disponibilidad Biológica , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Portadoras/metabolismo , Bovinos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Coristoma/patología , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Medios de Cultivo Condicionados/farmacología , Disacáridos/metabolismo , Sinergismo Farmacológico , Femenino , Heparina/farmacología , Humanos , Ratones , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Sus scrofa
7.
J Biol Chem ; 286(20): 17755-65, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454472

RESUMEN

We compare here the structural and functional properties of heparan sulfate (HS) chains from both male or female adult mouse liver through a combination of molecular sieving, enzymatic cleavage, and strong anion exchange-HPLC. The results demonstrated that male and female HS chains are significantly different by a number of parameters; size determination showed that HS chain lengths were ∼100 and ∼22 kDa, comprising 30-40 and 6-8 disaccharide repeats, respectively. Enzymatic depolymerization and disaccharide composition analyses also demonstrated significant differences in domain organization and fine structure. N-Unsubstituted glucosamine (ΔHexA-GlcNH(3)(+), ΔHexA-GlcNH(3)(+)(6S), ΔHexA(2S)-GlcNH(3)(+), and N-acetylglucosamine (ΔHexA-GlcNAc) are the predominant disaccharides in male mouse liver HS. However, N-sulfated glucosamine (ΔHexA-GlcNSO(3)) is the predominant disaccharide found in female liver. These structurally different male and female liver HS forms exert differential effects on human mesenchymal cell proliferation and subsequent osteogenic differentiation. The present study demonstrates the potential usefulness of gender-specific liver HS for the manipulation of human mesenchymal cell properties, including expansion, multipotentiality, and subsequent matrix mineralization. Our results suggest that HS chains show both tissue- and gender-specific differences in biochemical composition that directly reflect their biological activity.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/farmacología , Hígado/química , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Caracteres Sexuales , Adulto , Animales , Femenino , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos CBA
8.
Pharm Res ; 28(6): 1385-94, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21184148

RESUMEN

PURPOSE: In order to address cell dose limitations associated with the use of cord blood hematopoietic stem cell (HSC) transplantation, we explored the effect of bone marrow stroma-derived heparan sulfate (HS) on the ex vivo expansion of HSCs. METHODS: Heparan sulfate was isolated and purified from the conditioned media of human bone marrow stromal cells and used for the expansion of cord blood-derived CD34(+) cells in the presence of a cocktail of cytokines. RESULTS: The number of myeloid lineage-committed progenitor cells was increased at low dosage of HS as illustrated by an increase in the total number of colony-forming cells (CFC) and colonies of erythroid (BFU-E) and granulocyte-macrophage (CFU-GM) precursors. Notably, the stroma-derived HS did not alter the growth of CD34(+) HSCs or negatively affect the levels of various HSC phenotypic markers after expansion. CONCLUSIONS: This study shows that HS secreted into solution by stromal cells has the capacity to support hematopoietic cytokines in the maintenance and expansion of HSCs. The incorporation of stroma-derived HS as a reagent may improve the efficacy of cord blood HSC transplantation by enhancing the number of committed cells and accelerating the rate of engraftment.


Asunto(s)
Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Heparitina Sulfato/farmacología , Células de la Médula Ósea/química , Recuento de Células , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Medios de Cultivo Condicionados , Glicosaminoglicanos/química , Glicosaminoglicanos/aislamiento & purificación , Glicosaminoglicanos/farmacología , Células Progenitoras de Granulocitos y Macrófagos/citología , Células Progenitoras de Granulocitos y Macrófagos/efectos de los fármacos , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Humanos , Células Progenitoras de Megacariocitos y Eritrocitos/citología , Células Progenitoras de Megacariocitos y Eritrocitos/efectos de los fármacos , Células Madre , Células del Estroma/química
9.
J Cell Biochem ; 109(6): 1222-31, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20135643

RESUMEN

Skeletal integrity is tightly regulated by the activity of osteoblasts and osteoclasts that are both under the control of extracellular glycosaminoglycans (GAGs) through their interactions with endogenous growth factors and differentiation-promoting ligands. Receptor activator of NF-kappa-B ligand (RANKL), which is a tumor necrosis factor (TNF)-related protein that is critical for osteoclast formation, is produced by osteoblasts and further modulated by certain types of GAGs. Using unfractionated osteoblast-derived GAGs that reflect the complex tissue microenvironment within which osteoclasts reside, we demonstrate that these GAGs block the osteoclastogenic activity of RANKL. Furthermore, RANKL significantly reduces extracellular signal-regulated protein kinase (ERK) activity, a putative suppressor of osteoclastogenesis, but osteoblast-derived GAGs eliminate the inhibitory effects of RANKL on ERK activity. Notably, while imposing an anti-osteoclastic effect, these GAGs also enhanced the proliferation of osteoblasts. Thus, the osteoblast microenvironment is a potent source of GAGs that promote bone anabolic activities. The anti-osteoclastogenic and osteoblast-related mitogenic activities of these GAGs together may provide a key starting point for the development of selective sugar-based therapeutic compounds for the treatment of osteopenic disorders.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/farmacología , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Ligando RANK/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Cromatografía por Intercambio Iónico , Immunoblotting , Ratones , Microscopía Confocal , Osteoclastos/metabolismo , Porcinos
10.
J Cell Biochem ; 108(5): 1132-42, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19777445

RESUMEN

Heparan sulfate (HS) is a linear, highly variable, highly sulfated glycosaminoglycan sugar whose biological activity largely depends on internal sulfated domains that mediate specific binding to an extensive range of proteins. In this study we employed anion exchange chromatography, molecular sieving and enzymatic cleavage on HS fractions purified from three compartments of cultured osteoblasts-soluble conditioned media, cell surface, and extracellular matrix (ECM). We demonstrate that the composition of HS chains purified from the different compartments is structurally non-identical by a number of parameters, and that these differences have significant ramifications for their ligand-binding properties. The HS chains purified of conditioned medium had twice the binding affinity for FGF2 when compared with either cell surface or ECM HS. In contrast, similar binding of BMP2 to the three types of HS was observed. These results suggest that different biological compartments of cultured cells have structurally and functionally distinct HS species that help to modulate the flow of HS-dependent factors between the ECM and the cell surface.


Asunto(s)
Heparitina Sulfato/análisis , Heparitina Sulfato/aislamiento & purificación , Osteoblastos/química , Proteína Morfogenética Ósea 2/química , Proteína Morfogenética Ósea 2/metabolismo , Membrana Celular/química , Células Cultivadas , Medios de Cultivo Condicionados/química , Matriz Extracelular/química , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Humanos , Osteoblastos/metabolismo , Osteoblastos/ultraestructura , Unión Proteica
11.
J Cell Physiol ; 220(3): 780-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19479939

RESUMEN

Heparan sulfate (HS) sugar chains attached to core proteoglycans (PGs) termed HSPGs mediate an extensive range of cell-extracellular matrix (ECM) and growth factor interactions based upon their sulfation patterns. When compared with non-osteogenic (maintenance media) culture conditions, under established osteogenic culture conditions, MC3T3-E1 cells characteristically increase their osteogenic gene expression profile and switch their dominant fibroblast growth factor receptor (FGFR) from FGFR1 (0.5-fold decrease) to FGFR3 (1.5-fold increase). The change in FGFR expression profile of the osteogenic-committed cultures was reflected by their inability to sustain an FGF-2 stimulus, but respond to BMP-2 at day 14 of culture. The osteogenic cultures decreased their chondroitin and dermatan sulfate PGs (biglycan, decorin, and versican), but increased levels of the HS core protein gene expression, in particular glypican-3. Commitment and progress through osteogenesis is accompanied by changes in FGFR expression, decreased GAG initiation but increased N- and O-sulfation and reduced remodeling of the ECM (decreased heparanase expression) resulting in the production of homogenous (21 kDa) HS chain. With the HSPG glypican-3 expression strongly upregulated in these processes, siRNA was used to knockdown this gene to examine the effect on osteogenic commitment. Reduced glypican-3 abrogated the expression of Runx2, and thus differentiation. The reintroduction of this HSPG into Runx2-null cells allowed osteogenesis to proceed. These results demonstrate the dependence of osteogenesis on specific HS chains, in particular those associated with glypican-3.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Glipicanos/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Procesamiento Proteico-Postraduccional , Células 3T3 , Animales , Biglicano , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Decorina , Proteínas de la Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica , Glipicanos/genética , Liasa de Heparina/genética , Liasa de Heparina/metabolismo , Humanos , Ratones , Ratones Noqueados , Osteoblastos/enzimología , Osteogénesis/genética , Proteoglicanos/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Factores de Tiempo , Transfección , Versicanos/metabolismo
12.
J Mol Histol ; 38(5): 469-81, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17674130

RESUMEN

Heparan sulfate proteoglycans (HSPGs) are complex and labile macromolecular moieties on the surfaces of cells that control the activities of a range of extracellular proteins, particularly those driving growth and regeneration. Here, we examine the biosynthesis of heparan sulfate (HS) sugars produced by cultured MC3T3-E1 mouse calvarial pre-osteoblast cells in order to explore the idea that changes in HS activity in turn drive phenotypic development during osteogenesis. Cells grown for 5 days under proliferating conditions were compared to cells grown for 20 days under mineralizing conditions with respect to their phenotype, the forms of HS core protein produced, and their HS sulfotransferase biosynthetic enzyme levels. RQ-PCR data was supported by the results from the purification of day 5 and day 20 HS forms by anionic exchange chromatography. The data show that cells in active growth phases produce more complex forms of sugar than cells that have become relatively quiescent during active mineralization, and that these in turn can differentially influence rates of cell growth when added exogenously back to preosteoblasts.


Asunto(s)
Glicosaminoglicanos/metabolismo , Osteogénesis/fisiología , Sulfotransferasas/metabolismo , Células 3T3 , Animales , Calcificación Fisiológica/genética , Calcificación Fisiológica/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Glucuronidasa/genética , Glucuronidasa/metabolismo , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Heparitina Sulfato/aislamiento & purificación , Heparitina Sulfato/metabolismo , Heparitina Sulfato/farmacología , Immunoblotting , Ratones , Osteogénesis/genética , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfotransferasas/genética
13.
J Cell Physiol ; 210(1): 38-50, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17051597

RESUMEN

The transcription factor Runx2 can be controlled by a number of upstream regulators involved in intracellular signalling, including the activation ERK1/2 signaling by fibroblast growth factor-2 (FGF-2). FGFs interact with their cell surface receptors (FGFRs) through an obligate cross-binding interaction with heparan sulfate proteoglycan (HSPG) co-receptors; exogenous HS sugar chains have been shown to potently modulate changes in cell phenotype depending on the stage of tissue differentiation when the HS is harvested, suggesting that HS chain structure and function varies depending on the stage of cell maturity. This study examined the potential of bone-derived heparan sulfate (HS), harvested from differentiating osteoblasts, for the enhancement of preosteoblast growth and differentiation. HS was harvested from conditioned media, cell surface and matrix compartments of postconfluent (differentiating) MC3T3-E1 osteoblasts and dosed back onto preconfluent MC3T3-E1 cells. We show that HS can increase the expression Runx2, ALP, and OPN in preosteoblast cells, suggesting the potential for exogenous HS to shift cells from proliferative to differentiative phenotypes. In line with their structural differences, only HS released into the media was found to co-stimulate the mitogenic effect of FGF-2, whilst exogenous application of all the HSs together with FGF-2 served to increase the expression of OPN. Only the application of cell surface-derived HS triggered a synergistic increase in FGFR1 expression together with FGF-2, although all three HS preparations could trigger transient increases in PI3K, ERK1/2, and stat3 phosphorylation levels. These findings demonstrate that the compartmentally distinct HS species expressed by differentiating MC3T3-E1 cells act in complex ways to coordinate the extracellular conditions that lead to osteoblast differentiation, with the cell surface species coordinating the FGF response.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Heparitina Sulfato/farmacología , Osteoblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Animales , Butadienos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/química , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Nitrilos/farmacología , Osteoblastos/química , Osteoblastos/citología , Osteoblastos/metabolismo , Osteopontina/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo
14.
J Cell Physiol ; 209(3): 811-25, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16972247

RESUMEN

Fibroblast growth factor-2 (FGF2) is a powerful promoter of bone growth. We demonstrate here that brief exposure to FGF2 enhances mineralized nodule formation in cultured rat osteoprogenitor cells due to an expansion of cells that subsequently mineralize. This mitogenic effect is mediated via sulfated glycosaminoglycans (GAGs), FGFR1, and the extracellular signal-regulated kinase (ERK) pathway. The GAGs involved in this stimulation are chondroitin sulfates (CS) rather than heparan sulfates (HS). However, continuous FGF2 treatment reduces alkaline phosphatase (ALP) activity, downregulates collagen Ialpha1 (ColIalpha1) and FGFR3 expression, upregulates the expression and secretion of osteopontin (OPN) and inhibits mineralization. The inhibitory effects of FGF2 on FGFR3 expression and ALP activity are also mediated by the ERK pathway, although the effects of FGF2 on ColIalpha1 and OPN expression are mediated by GAGs and PKC activity. Thus short-term activation of FGF2/FGFR1 promotes osteoprogenitor proliferation and subsequent differentiation, while long-term activation of FGF2 signaling disrupts mineralization by modulating osteogenic marker expression. This study thus establishes the central role of sulfated GAGs in the osteogenic progression of osteoprogenitors.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glicosaminoglicanos , Osteogénesis/fisiología , Cráneo/citología , Células Madre/fisiología , Sulfatos/química , Fosfatasa Alcalina/metabolismo , Animales , Calcificación Fisiológica/fisiología , Proliferación Celular , Células Cultivadas , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Osteopontina/metabolismo , Ratas , Ratas Wistar , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...