Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38740375

ABSTRACT

INTRODUCTION: We previously showed that a 3-week oral metformin (MET) treatment enhances the osteogenic potential of bone marrow stromal cells (BMSCs) and improves several bone histomorphometric parameters in Wistar rats with metabolic syndrome (MetS). However, the skeletal effects of extended periods of MET need to be completely elucidated. Hence, in this study, the impact of a prolonged (3-month) MET treatment was investigated on bone architecture, histomorphometric and biomechanics variables, and osteogenic potential of BMSCs in Wistar rats with or without MetS. MATERIALS AND METHODS: Young male Wistar rats (n=36) were randomized into four groups (n=9) that received either 20% fructose (F), MET (MET), F plus MET treatments (FMET), or drinking water alone (Veh). Rats were euthanized, blood was collected, and bones were dissected and processed for peripheral quantitative computed tomography (pQCT) analysis, static and dynamic histomorphometry, and bone biomechanics. In addition, BMSCs were isolated to determine their osteogenic potential. RESULTS: MET affected trabecular and cortical bone, altering bone architecture and biomechanics. Furthermore, MET increased the pro-resorptive profile of BMSCs. In addition, fructose-induced MetS practically did not affect the the structural or mechanical variables of the skeleton. CONCLUSION: A 3-month treatment with MET (with or without MetS) affects bone architecture and biomechanical variables in Wistar rats.

2.
Actual. osteol ; 18(3): 169-182, 2022. ilus, tab
Article in Spanish | LILACS, UNISALUD, BINACIS | ID: biblio-1509315

ABSTRACT

El síndrome metabólico se define como un trastorno heterogéneo y multifactorial con riesgo cardiovascular elevado. Actualmente se encuentra en franco crecimiento debido al sedentarismo y la ingesta rica en grasas y azúcares. Su tratamiento incluye la indicación de cambios en el estilo de vida, con realización de actividad física y una alimentación saludable e hipocalórica. Cuando esto no es eficaz, se pueden utilizar diferentes fármacos, y entre los más prescriptos se encuentra la metformina, caracterizada por su acción insulino-sensibilizante. Numerosos trabajos han estudiado la vinculación del síndrome metabólico con el tejido óseo. Se demostró como resultado general, aunque no concluyente, que dicho síndrome se asocia con una disminución de la densidad mineral ósea y un aumento en la incidencia de fracturas osteoporóticas. Una de las limitaciones de estos estudios clínicos estaría ligada a la gran heterogeneidad de los pacientes con síndrome metabólico. Por otra parte, y dado que diversos estudios preclínicos han sugerido posibles acciones osteogénicas de la metformina, se ha investigado el posible efecto óseo de un tratamiento con este fármaco en personas con hiperglucemia o disglucemia. Varios estudios clínicos muestran que este efecto sería nulo o, en algunos casos, de carácter protector para el sistema óseo. No obstante, se debería tener precaución en el uso de dicho fármaco en pacientes que necesiten dosis altas y/o posean riesgo elevado de fractura, ya que sus altas concentraciones podrían tener consecuencias negativas sobre el metabolismo óseo. (AU)


Metabolic syndrome is defined as a heterogeneous and multifactorial disorder with high cardiovascular risk. Its incidence is currently growing due to sedentary lifestyles and diets with a high intake of fats and sugars. Treatment for metabolic syndrome begins with changes in lifestyle, such as physical activity and a healthy and hypocaloric diet. When this is not effective, different drugs can be used, and one of the most frequently prescribed is the insulin-sensitizer metformin. Numerous investigations have evaluated the possible link between metabolic syndrome and alterations in bone metabolism. Although not conclusive, most clinical studies point to an association between metabolic syndrome, a decrease in bone mineral density and an increase in the incidence of osteoporotic fractures. However, an important limitation of these studies is the great heterogeneity of individuals with metabolic syndrome. In view of preclinical research indicating possible osteogenic actions of metformin, the effects on bone of metformin has been evaluated in patients with hyperglycemia. Most studies have found either no effect on fracture incidence, or a mild protective action. However, since elevated concentrations of metformin might negatively affect bone metabolism, caution should be taken when prescribing this drug for patients who require high doses, and/or have an excess fracture risk. (AU)


Subject(s)
Humans , Bone and Bones/drug effects , Metabolic Syndrome/drug therapy , Metformin/administration & dosage , Bone Diseases, Metabolic/complications , Bone Density , Metabolic Syndrome/physiopathology , Fractures, Bone/epidemiology , Metformin/pharmacology
3.
J Biomater Sci Polym Ed ; 21(10): 1297-312, 2010.
Article in English | MEDLINE | ID: mdl-20534186

ABSTRACT

There is considerable interest in the design of polymeric biomaterials that can be used for the repair of bone defects. In this study, we used ultrasound to prepare a compatibilized blend of poly(epsilon-caprolactone) (PCL) and poly(diisopropyl fumarate) (PDIPF). The formation of post-sonication inter-polymer coupling products was verified by SEC analysis of a blend with azo-labeled PDIPF. We also analyzed the physicochemical and mechanical properties of the compatibilized blend. When compared to PCL alone, the PCL/PDIPF blend showed no difference in its resistance as evaluated by the elastic modulus, although it did show a 50% decrease in ultimate tensile stress (P < 0.05) and an 84% decrease in elongation-at-break (P < 0.05). However, the mechanical properties of this blend were comparable to those of trabecular bone. We next evaluated biocompatibility of the PCL/PDIPF blend, and of homo-polymeric PCL and PDIPF films for comparison, with UMR106 and MC3T3E1 osteoblastic cells. Osteoblasts plated on the compatibilized blend adhered and proliferated more than on either homo-polymer, showed a greater number of cellular processes with a better organized actin cytoskeleton and expressed more type-I collagen and mineral, both markers of osteoblast phenotype. These results support the hypothesis that this new compatibilized blend could be useful in future applications for bone regeneration.


Subject(s)
Biocompatible Materials/chemistry , Bone and Bones , Fumarates/chemistry , Polyesters/chemistry , Polymers/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/adverse effects , Cell Adhesion , Cell Line , Cell Line, Tumor , Cell Proliferation , Materials Testing , Mice , Microscopy, Electron, Scanning , Polymers/adverse effects , Polymers/chemical synthesis , Rats , Tissue Scaffolds/adverse effects
4.
J Bone Miner Res ; 25(2): 211-21, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19594306

ABSTRACT

Diabetes mellitus is associated with bone loss. Patients with type 2 diabetes are frequently treated with oral antidiabetic drugs such as sulfonylureas, biguanides, and thiazolidinediones. Rosiglitazone treatment has been shown to increase adipogenesis in bone marrow and to induce bone loss. In this study we evaluated the effect of in vivo and in vitro treatment with metformin on bone marrow progenitor cells (BMPCs), as well as the involvement of AMPK pathway in its effects. The in vitro effect of coincubation with metformin and rosiglitazone on the adipogenic differentiation of BMPCs also was studied. In addition, we evaluated the effect of in vivo metformin treatment on bone regeneration in a model of parietal lesions in nondiabetic and streptozotocin-induced diabetic rats. We found that metformin administration both in vivo and in vitro caused an increase in alkaline phosphatase activity, type I collagen synthesis, osteocalcin expression, and extracellular calcium deposition of BMPCs. Moreover, metformin significantly activated AMPK in undifferentiated BMPCs. In vivo, metformin administration enhanced the expression of osteoblast-specific transcription factor Runx2/Cbfa1 and activation of AMPK in a time-dependent manner. Metformin treatment also stimulated bone lesion regeneration in control and diabetic rats. In vitro, metformin partially inhibited the adipogenic actions of rosiglitazone on BMPCs. In conclusion, our results indicate that metformin causes an osteogenic effect both in vivo and in vitro, possibly mediated by Runx2/Cbfa1 and AMPK activation, suggesting a possible action of metformin in a shift toward the osteoblastic differentiation of BMPCs.


Subject(s)
Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Stem Cells/drug effects , Animals , Blotting, Western , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Enzyme Activation/drug effects , Fibrinolytic Agents/pharmacokinetics , Male , Osteoblasts/drug effects , Rats , Rats, Sprague-Dawley , Rosiglitazone , Thiazolidinediones/pharmacology
5.
Eur J Pharmacol ; 536(1-2): 38-46, 2006 Apr 24.
Article in English | MEDLINE | ID: mdl-16564524

ABSTRACT

An association has been previously established between uncompensated diabetes mellitus and the loss of bone mineral density and/or quality. In this study, we evaluated the effects of metformin on the growth and differentiation of osteoblasts in culture. Treatment of two osteoblast-like cells (UMR106 and MC3T3E1) with metformin (25-500 microM) for 24 h led to a dose-dependent increase of cell proliferation. Metformin also promoted osteoblastic differentiation: it increased type-I collagen production in both cell lines and stimulated alkaline phosphatase activity in MC3T3E1 osteoblasts. In addition, metformin markedly increased the formation of nodules of mineralization in 3-week MC3T3E1 cultures. Metformin induced activation and redistribution of phosphorylated extracellular signal-regulated kinase (P-ERK) in a transient manner, and dose-dependently stimulated the expression of endothelial and inducible nitric oxide synthases (e/iNOS). These results show for the first time a direct osteogenic effect of metformin on osteoblasts in culture, which could be mediated by activation/redistribution of ERK-1/2 and induction of e/iNOS.


Subject(s)
Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Osteoblasts/drug effects , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen Type I/biosynthesis , Dose-Response Relationship, Drug , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , Phosphorylation/drug effects
6.
Biol Trace Elem Res ; 104(2): 131-40, 2005 May.
Article in English | MEDLINE | ID: mdl-15894813

ABSTRACT

Bisphosphonates (BPs) are drugs widely used in the treatment of various bone diseases. BPs localize to bone mineral, and their concentration in resorption lacunae could reach almost millimolar levels. Bone alkaline phosphatase (ALP) is a membrane-bound exoenzyme that has been implicated in bone formation and mineralization. In this study, we investigated the possible direct effect of three N-containing BPs (alendronate, pamidronate, and zoledronate) on the specific activity of bone ALP obtained from an extract of UMR106 rat osteosarcoma cells. Enzymatic activity was measured by spectrophotometric detection of p-nitrophenol product and by in situ visualization of ALP bands after an electrophoresis on cellulose acetate gels. Because ALP is a metalloprotein that contains Zn2+ and Mg2+, both of which are necessary for catalytic function, we also evaluated the participation of these divalent cations in the possible effect of BPs on enzymatic activity. All BPs tested were found to dose-dependently inhibit spectrophotometrically measured ALP activity (93-42% of basal) at concentrations of BPs between 10-5 M and 10-4 M, the order of potency being zoledronate approximately equals alendronate > pamidronate. However, coincubation with excess Zn2+ or Mg2+ completely abolished this inhibitory effect. Electrophoretic analysis rendered very similar results: namely a decrease in the enzymatic activity of the bone-ALP band by BPs and a reversion of this inhibition by divalent cations. This study shows that N-containing BPs directly inhibit bone-ALP activity, in a concentration range to which this exoenzyme is probably exposed in vivo. In addition, this inhibitory effect is most possibly the result of the chelation of Zn2+ and Mg2+ ions by BPs.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Bone and Bones/enzymology , Cations, Divalent/pharmacology , Diphosphonates/pharmacology , Magnesium/pharmacology , Zinc/pharmacology , Alendronate/pharmacology , Animals , Bone Resorption/drug therapy , Imidazoles/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/isolation & purification , Osteoblasts/enzymology , Osteosarcoma/enzymology , Pamidronate , Rats , Tumor Cells, Cultured , Zoledronic Acid
7.
Mol Cell Biochem ; 266(1-2): 17-24, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15646023

ABSTRACT

The accumulation of irreversible advanced glycation endproducts (AGEs) on long-lived proteins, and the interaction of AGEs with cellular receptors such as AGE-R3/galectin-3 and RAGE, are considered to be key events in the development of long-term complications of diabetes mellitus, Alzheimer's disease, uremia and ageing. The aim of this study was to investigate the expression and sub-cellular distribution of galectin-3, as well as its possible modulation by AGEs, in MC3T3E1 mouse calvaria-derived osteoblasts and in UMR 106 rat osteosarcoma cells. Both osteoblastic lines were cultured either with control bovine serum albumin (BSA) or with AGEs-BSA for 48 h. Cells were evaluated for galectin-3 expression by fixing and immunofluorescent microscopic analysis; or Western blot analysis of whole cell extracts, sub-cellular fractions and culture media. Both cell lines express 30 kDa (monomeric) galectin-3, although expression was about 15-fold lower in the UMR106 osteosarcoma cells. Dimeric (70 kDa) galectin-3 was additionally observed in the UMR106 cells. Immunofluorescent analysis of galectin-3 distribution showed a diffuse cytoplasmic and strong nuclear pattern in MC3T3E1 osteoblasts, and a patchy cytoplasmic pattern in UMR106 cells. Western blot analysis for both cell lines showed that galectin-3 was mainly found in the cytoplasm and in minor amounts in the microsomal fraction, while considerable amounts were secreted into the culture media. Exposure to 100-200 microg/mL AGEs-BSA increased the cellular content of 30 kDa galectin-3 (20-25% for MC3T3E1 and 35-70% for UMR106 versus control BSA, p < 0.05), and decreased the culture media levels of galectin-3 (10-20% for MC3T3E1 and for UMR106 versus control BSA, p < 0.05). These results confirm the expression of galectin-3 in osteoblastic cells, and suggest different levels and sub-cellular distribution of this protein in transformed versus non-transformed osteoblasts. Osteoblastic exposure to AGEs alters their expression and secretion of galectin-3, which could have significant consequences on osteoblast metabolism and thus on bone turnover.


Subject(s)
Galectin 3/metabolism , Glycation End Products, Advanced/pharmacology , Osteoblasts/metabolism , Serum Albumin, Bovine/pharmacology , Aging/metabolism , Alzheimer Disease/metabolism , Animals , Cattle , Cell Line, Transformed , Cell Transformation, Neoplastic/metabolism , Diabetes Mellitus/metabolism , Gene Expression Regulation/drug effects , Mice , Rats , Receptor for Advanced Glycation End Products , Receptors, Immunologic , Signal Transduction/drug effects , Uremia/metabolism
8.
Mol Cell Biochem ; 250(1-2): 1-10, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12962137

ABSTRACT

An increase in the interaction between advanced glycation end-products (AGEs) and their receptor RAGE is believed to contribute to the pathogenesis of chronic complications of Diabetes mellitus, which can include bone alterations such as osteopenia. We have recently found that extracellular AGEs can directly regulate the growth and development of rat osteosarcoma UMR106 cells, and of mouse calvaria-derived MC3T3E1 osteoblasts throughout their successive developmental stages (proliferation, differentiation and mineralisation), possibly by the recognition of AGEs moieties by specific osteoblastic receptors which are present in both cell lines. In the present study we examined the possible expression of RAGE by UMR106 and MC3T3E1 osteoblastic cells, by immunoblot analysis. We also investigated whether short-, medium- or long-term exposure of osteoblasts to extracellular AGEs, could modify their affinity constant and maximal binding for AGEs (by 125I-AGE-BSA binding experiments), their expression of RAGE (by immunoblot analysis) and the activation status of the osteoblastic ERK 1/2 signal transduction mechanism (by immunoblot analysis for ERK and P-ERK). Our results show that both osteoblastic cell lines express readily detectable levels of RAGE. Short-term exposure of phenotypically mature osteoblastic UMR106 cells to AGEs decrease the cellular density of AGE-binding sites while increasing the affinity of these sites for AGEs. This culture condition also dose-dependently increased the expression of RAGE and the activation of ERK. In proliferating MC3T3E1 pre-osteoblasts, 24-72 h exposure to AGEs did not modify expression of RAGE, ERK activation or the cellular density of AGE-binding sites. However, it did change the affinity of these binding sites forAGEs, with both higher- and lower-affinity sites now being apparent. Medium-term ( 1 week) incubation of differentiated MC3T3E1 osteoblasts with AGEs, induced a simultaneous increase in RAGE expression and in the relative amount of P-ERK. Mineralising MC3T3E1 cultures grown for 3 weeks in the presence of extracellular AGEs showed a decrease both in RAGE and P-ERK expression. These results indicate that, in phenotypically mature osteoblastic cells, changes in ERK activation closely follow the AGEs-induced regulation of RAGE expression. Thus, the AGEs-induced biological effects that we have observed previously in osteoblasts, could be mediated by RAGE in the later stages of development, and mediated by other AGE receptors in the earlier pre-osteoblastic stage.


Subject(s)
Glycation End Products, Advanced/physiology , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/metabolism , 3T3 Cells , Animals , Binding Sites , Blotting, Western , Cell Differentiation , Cell Division , Cell Line, Tumor , Collagen Type I/chemistry , Dose-Response Relationship, Drug , Enzyme Activation , Immunoblotting , Kinetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Phenotype , Protein Binding , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...