Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Diabetes Complications ; 37(11): 108626, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37839167

RESUMO

AIMS: Our objective was to study the vascular smooth muscle cells (VSMC) osteoblastic transdifferentiation in AGE exposed cells or those from diabetic animals, and its response to metformin treatment. METHODS: VSMC were obtained from non-diabetic rats, grown with or without AGE; while VSMC of in vivo-ex vivo studies were obtained from non-diabetic control animals (C), diabetic (D), C treated with metformin (M) and D treated with metformin (D-M). We studied the osteoblastic differentiation by evaluating alkaline phosphatase (ALP), type I collagen (Col) and mineral deposit. RESULTS: In vitro, AGE increased proliferation, migration, and osteoblastic differentiation of VSMC. Metformin cotreatment prevented the AGE induced proliferation and migration. Both AGE and metformin stimulated the expression of ALP and Col. AGE induced mineralization was prevented by metformin. VSMC from D expressed a higher production of Col and ALP. Those from D-M showed an ALP increase vs C and M, and a partial decrease vs D. Cultured in osteogenic medium, ALP, Col and mineralization increased in D vs C, remained unchanged in M, and were prevented in D-M animals. CONCLUSION: Both AGE and DM favor VSMC differentiation towards the osteogenic phenotype and this effect can be prevented by metformin.


Assuntos
Calcinose , Diabetes Mellitus , Calcificação Vascular , Ratos , Animais , Produtos Finais de Glicação Avançada/metabolismo , Músculo Liso Vascular/metabolismo , Transdiferenciação Celular , Reação de Maillard , Diabetes Mellitus/metabolismo , Células Cultivadas
2.
Biol Trace Elem Res ; 186(2): 457-466, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29623650

RESUMO

Long-term diabetes mellitus can induce osteopenia and osteoporosis, an increase in the incidence of low-stress fractures, and/or delayed fracture healing. Strontium ranelate (SrR) is a dual-action anti-osteoporotic agent whose use in individuals with diabetic osteopathy has not been adequately evaluated. In this study, we studied the effects of an oral treatment with SrR and/or experimental diabetes on bone composition and biomechanics. Young male Wistar rats (half non-diabetic, half with streptozotocin/nicotinamide-induced diabetes) were either untreated or orally administered 625 mg/kg/day of SrR for 6 weeks. After sacrifice, femora from all animals were evaluated by a multi-scale approach (X-ray diffraction, Fourier transform infrared spectroscopy, inductively coupled plasma optical-emission spectrometry, static histomorphometry, pQCT, and mechanical testing) to determine chemical, crystalline, and biomechanical properties. Untreated diabetic animals (versus untreated non-diabetic) showed a decrease in femoral mineral carbonate content, in cortical thickness and BMC, in trabecular osteocyte density, in maximum load supported at rupture and at yield point, and in overall toughness at mid-shaft. Treatment of diabetic animals with SrR further affected several parameters of bone (some already impaired by diabetes): crystallinity index (indicating less mature apatite crystals); trabecular area, BMC, and vBMD; maximum load at yield point; and structural elastic rigidity. However, SrR was also able to prevent the diabetes-induced decreases in trabecular osteocyte density (completely) and in bone ultimate strength at rupture (partially). Our results indicate that SrR treatment can partially but significantly prevent some bone structural mechanical properties as previously affected by diabetes, but not others (which may even be worsened).


Assuntos
Densidade Óssea/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Diabetes Mellitus Experimental/fisiopatologia , Tiofenos/farmacologia , Administração Oral , Animais , Densidade Óssea/fisiologia , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/farmacologia , Doenças Ósseas/patologia , Doenças Ósseas/fisiopatologia , Doenças Ósseas/prevenção & controle , Fêmur/citologia , Fêmur/efeitos dos fármacos , Fêmur/fisiologia , Masculino , Osteócitos/citologia , Osteócitos/efeitos dos fármacos , Ratos Wistar , Tiofenos/administração & dosagem
3.
J Biomed Mater Res A ; 106(2): 570-579, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28984066

RESUMO

In the tissue engineering field, the design of the scaffold inspired on the natural occurring tissue is of vital importance. Ideally, the scaffold surface must promote cell growth and differentiation, while promote angiogenesis in the in vivo implant of the scaffold. On the other hand, the material selection must be biocompatible and the degradation times should meet tissue reparation times. In the present work, we developed a nanofibrous scaffold based on chitosan crosslinked with diisopropylfumarate-vinyl acetate copolymer using anodized aluminum oxide (AAO) templates. We have previously demonstrated its biocompatibility properties with low cytotoxicity and proper degradation times. Now, we extended our studies to demonstrate that it can be successfully nanostructured using the AAO templates methodology, obtaining a nanorod-like scaffold with a diameter comparable to those of collagen fibers of the bone matrix (170 and 300 nm). The nanorods obtained presented a very homogeneous pattern in diameter and length, and supports cell attachment and growth. We also found that both osteoblastic and chondroblastic matrix production were promoted on bone marrow progenitor cells and primary condrocytes growing on the scaffolds, respectively. In addition, the nanostructured scaffold presented no cytotoxicity as it was evaluated using a model of macrophages on culture. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 570-579, 2018.


Assuntos
Quitosana/química , Condrogênese , Fumaratos/química , Nanoestruturas/química , Osteogênese , Polímeros/química , Regeneração , Alicerces Teciduais/química , Óxido de Alumínio/química , Animais , Materiais Biocompatíveis/química , Morte Celular , Reagentes de Ligações Cruzadas/química , Eletrodos , Camundongos , Nanofibras/química , Nanofibras/ultraestrutura , Óxido Nítrico/metabolismo , Polímeros/síntese química , Células RAW 264.7 , Ratos Sprague-Dawley , Análise Espectral Raman , Termogravimetria , Água/química
4.
Mol Cell Endocrinol ; 450: 94-104, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28456475

RESUMO

Advanced glycation end products (AGE) have been demonstrated to induce the osteogenic trans-differentiation of vascular smooth muscle cells (VSMC). Strontium ranelate (SR) is an anti-osteoporotic agent that has both anti-catabolic and anabolic actions on bone tissue. However, in the last years SR has been associated with an increase of cardiovascular risk. We hypothesize that SR can increase the osteoblastic trans-differentiation of VSMC and the induction of extracellular calcifications, an effect that could be potentiated in the presence of AGE and inhibited by simultaneous administration of vitamin D. The present results of our in vitro experiments demonstrate that AGE and SR alone or in combination, stimulate L-type calcium channels, causing an increase in reactive oxygen species and activation of both ERK and NFkB, with the final effect of promoting the osteogenic shift of VSMC. Importantly, these in vitro effects of AGE and/or SR can be prevented by co-incubation with vitamin D.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Produtos Finais de Glicação Avançada/farmacologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Osteogênese/efeitos dos fármacos , Tiofenos/farmacologia , Vitamina D/farmacologia , Animais , Ácido Ascórbico/farmacologia , Contagem de Células , Movimento Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Masculino , Modelos Biológicos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Nifedipino/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Sulfassalazina/farmacologia , Vitamina E/farmacologia
5.
Macromol Biosci ; 17(5)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27892651

RESUMO

Natural and synthetic cross-linked polymers allow the improvement of cytocompatibility and mechanical properties of the individual polymers. In osteochondral lesions of big size it will be required the use of scaffolds to repair the lesion. In this work a borax cross-linked scaffold based on fumarate-vinyl acetate copolymer and chitosan directed to osteochondrondral tissue engineering is developed. The cross-linked scaffolds and physical blends of the polymers are analyzed in based on their morphology, glass transition temperature, and mechanical properties. In addition, the stability, degradation behavior, and the swelling kinetics are studied. The results demonstrate that the borax cross-linked scaffold exhibits hydrogel behavior with appropriated mechanical properties for bone and cartilage tissue regeneration. Bone marrow progenitor cells and primary chondrocytes are used to demonstrate its osteo- and chondrogenic properties, respectively, assessing the osteo- and chondroblastic growth and maturation, without evident signs of cytotoxicity as it is evaluated in an in vitro system.


Assuntos
Quitosana/química , Condrogênese , Fumaratos/química , Osteogênese , Polímeros/química , Engenharia Tecidual , Alicerces Teciduais , Animais , Biomarcadores/metabolismo , Células Cultivadas , Expressão Gênica , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Actual. osteol ; 12(2): 78-86, 2016. graf
Artigo em Espanhol | LILACS, UNISALUD, BINACIS | ID: biblio-1372017

RESUMO

La diabetes mellitus (DM) crónica se asocia con reducción en el contenido mineral óseo (osteopenia y osteoporosis). El objetivo de este trabajo fue evaluar la acción del ranelato de estroncio (RaSr) administrado por vía oral a animales control y diabéticos, sobre el potencial osteogénico de células progenitoras de médula ósea (CPMO). Dieciséis ratas Wistar macho jóvenes se dividieron en dos grupos: controles (C) y diabéticas (D) con destrucción parcial de células b-pancreáticas mediante inyecciones intraperitoneales consecutivas de nicotinamida y estreptozotocina. Siete días después de la inyección, cada grupo se subdividió: sin tratamiento, o tratadas oralmente con RaSr (625 mg/kg/día) durante seis semanas, luego de lo cual los animales fueron sacrificados. Las CPMO se obtuvieron de ratas de los cuatro grupos, por lavados del canal diafisario medular (húmero o fémur o ambos) y cultivo hasta confluencia en DMEM-10% FBS. La proliferación celular se evaluó mediante el ensayo de MTT. Luego las CPMO se replaquearon e incubaron en un medio osteogénico durante 14 días (fosfatasa alcalina [FAL] y colágeno tipo 1) o 21 días (mineralización). Las CPMO del grupo C+RaSr mostraron un aumento significativo versus control en la proliferación (133%) y en la diferenciación osteogénica (colágeno 143%, FAL 168%, mineralización 117%). La DM (grupo D) disminuyó significativamente la proliferación y diferenciación osteoblástica de las CPMO. El tratamiento con RaSr (grupo D+RaSr) previno completamente estos efectos antiosteogénicos de la DM. Así, en nuestro modelo experimental in vivo, la DM disminuye el potencial osteogénico de CPMO, efecto que puede ser prevenido por un tratamiento oral con RaSr. (AU)


Chronic diabetes mellitus (DM) is associated with a reduction in bone mineral content (osteopenia and osteoporosis). The object of this study was to evaluate the in vivo effect of he anti-osteoporotic drug strontium ranelate (SrRa) administered orally to control and diabetic animals, on the osteogenic potential of bone marrow progenitor cells (BMPC). Sixteen young male Wistar rats were divided into two groups: control (C) and diabetic with partial beta-cell destruction via consecutive intra-peritoneal injections of nicotinamide and streptozotocin (D). Seven days postinjection, each group was sub-divided: without treatment, or oral treatment with SrRa (625 mg/kg/day) for six weeks, after which the animals were euthanised (groups C, C+SrRa, D, D+SrRa). BMPC were obtained from rats of all four groups by flushing of the diaphysary canal (humerus and/or femur). Adherent cells were then cultured until confluence in DMEM10% FBS. Cell proliferation was evaluated with the MTT mitogenic bioassay. BMPC were replated and incubated in an osteogenic medium for 14 days (determination of alkaline phosphatase [ALP] and type-1 collagen) or 21 days (evaluation of mineralisation). BMPC from C+SrRa rats showed a significant increase versus control in proliferation (133%) and in osteogenic differentiation (collagen 143%, ALP 168%, mineralisation 117%). Induction of diabetes (group D) significantly decreased the proliferation and osteoblastic differentiation of BMPC. Treatment of diabetic animals with SrRa (group D+SrRa) completely prevented these anti-osteogenic effects of Diabetes. Thus, in our experimental in vivo model, Diabetes decreases the osteogenic potential of BMPC, an effect that can be prevented by oral treatment with strontium ranelate. (AU)


Assuntos
Animais , Masculino , Ratos , Osteoblastos/efeitos dos fármacos , Tiofenos/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Osteoporose/fisiopatologia , Tiofenos/administração & dosagem , Ratos Wistar , Modelos Animais de Doenças
7.
Biometals ; 27(3): 601-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24737106

RESUMO

Strontium ranelate (SR) is an orally administered and bone-targeting anti-osteoporotic agent that increases osteoblast-mediated bone formation while decreasing osteoclastic bone resorption, and thus reduces the risk of vertebral and femoral bone fractures in postmenopausal women with osteoporosis. Osteoblastic alkaline phosphatase (ALP) is a key enzyme involved in the process of bone formation and osteoid mineralization. In this study we investigated the direct effect of strontium (SR and SrCl2) on the activity of ALP obtained from UMR106 osteosarcoma cells, as well as its possible interactions with the divalent cations Zn(2+) and Mg(2+). In the presence of Mg(2+), both SR and SrCl2 (0.05-0.5 mM) significantly increased ALP activity (15-66 % above basal), and this was dose-dependent in the case of SR. The stimulatory effect of strontium disappeared in the absence of Mg(2+). The cofactor Zn(2+) also increased ALP activity (an effect that reached a plateau at 2 mM), and co-incubation of 2 mM Zn(2+) with 0.05-0.5 mM SR showed an additive effect on ALP activity stimulation. SR induced a dose-dependent decrease in the Km of ALP (and thus an increase in affinity for its substrate) with a maximal effect at 0.1 mM. Co-incubation with 2 mM Zn(2+) further decreased Km in all cases. These direct effects of SR on osteoblastic ALP activity could be indicating an alternative mechanism by which this compound may regulate bone matrix mineralization.


Assuntos
Fosfatase Alcalina/química , Conservadores da Densidade Óssea/química , Magnésio/química , Tiofenos/química , Zinco/química , Animais , Osso e Ossos/enzimologia , Linhagem Celular Tumoral , Hidrólise , Cinética , Nitrofenóis/química , Compostos Organofosforados/química , Ratos
8.
Eur J Pharmacol ; 727: 8-14, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24485890

RESUMO

Diabetes mellitus is associated with a decrease in bone quality and an increase in fracture incidence. Additionally, treatment with anti-diabetic drugs can either adversely or positively affect bone metabolism. In this study we evaluated: the effect of a 3-week oral treatment with saxagliptin on femoral microarchitecture in young male non-type-2-diabetic Sprague Dawley rats; and the in vitro effect of saxagliptin and/or fetal bovine serum (FBS), insulin or insulin-like growth factor-1 (IGF1), on the proliferation, differentiation (Runx2 and PPAR-gamma expression, type-1 collagen production, osteocalcin expression, mineralization) and extracellular-regulated kinase (ERK) activation, in bone marrow stromal cells (MSC) obtained from control (untreated) rats and in MC3T3E1 osteoblast-like cells. In vivo, oral saxagliptin treatment induced a significant decrease in the femoral osteocytic and osteoblastic density of metaphyseal trabecular bone and in the average height of the proximal cartilage growth plate; and an increase in osteoclastic tartrate-resistant acid phosphatase (TRAP) activity of the primary spongiosa. In vitro, saxagliptin inhibited FBS-, insulin- and IGF1-induced ERK phosphorylation and cell proliferation, in both MSC and MC3T3E1 preosteoblasts. In the absence of growth factors, saxagliptin had no effect on ERK activation or cell proliferation. In both MSC and MC3T3E1 cells, saxagliptin in the presence of FBS inhibited Runx2 and osteocalcin expression, type-1 collagen production and mineralization, while increasing PPAR-gamma expression. In conclusion, orally administered saxagliptin induced alterations in long-bone microarchitecture that could be related to its in vitro down-regulation of the ERK signaling pathway for insulin and IGF1 in MSC, thus decreasing the osteogenic potential of these cells.


Assuntos
Adamantano/análogos & derivados , Células da Medula Óssea/efeitos dos fármacos , Dipeptídeos/toxicidade , Inibidores da Dipeptidil Peptidase IV/toxicidade , Fêmur/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Adamantano/administração & dosagem , Adamantano/toxicidade , Administração Oral , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Proliferação de Células/efeitos dos fármacos , Dipeptídeos/administração & dosagem , Inibidores da Dipeptidil Peptidase IV/administração & dosagem , Relação Dose-Resposta a Droga , Fêmur/metabolismo , Fêmur/patologia , Masculino , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Fatores de Tempo
9.
Metabolism ; 63(2): 296-305, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24355623

RESUMO

OBJECTIVE: The aims of this study were: first, to evaluate the possible effects of a fructose rich diet (FRD)-induced metabolic syndrome (MS) on different aspects of long bone histomorphometry in young male rats; second, to investigate the effects of this diet on bone tissue regeneration; and third, to correlate these morphometric alterations with changes in the osteogenic/adipogenic potential and expression of specific transcription factors, of marrow stromal cells (MSC) isolated from rats with fructose-induced MS. MATERIALS/METHODS: MS was induced in rats by treatment with a FRD for 28 days. Halfway through treatment, a parietal wound was made and bone healing was evaluated 14 days later. After treatments, histomorphometric analysis was performed in dissected femoral and parietal bones. MSC were isolated from the femora of control or fructose-treated rats and differentiated either to osteoblasts (evaluated by type 1 collagen, Alkaline phosphatase and extracellular nodule mineralization) or to adipocytes (evaluated by intracellular triglyceride accumulation). Expression of Runx2 and PPARγ was assessed by Western blot. RESULTS: Fructose-induced MS induced deleterious effects on femoral metaphysis microarchitecture and impaired bone regeneration. Fructose treatment decreased the osteogenic potential of MSC and Runx2 expression. In addition, it increased the adipogenic commitment of MSC and PPARγ expression. CONCLUSIONS: Fructose-induced MS is associated with deleterious effects on bone microarchitecture and with a decrease in bone repair. These alterations could be due to a deviation in the adipogenic/osteogenic commitment of MSC, probably by modulation of the Runx2/PPARγ ratio.


Assuntos
Adipócitos/metabolismo , Células da Medula Óssea/metabolismo , Osso e Ossos/metabolismo , Frutose/administração & dosagem , Frutose/farmacologia , Síndrome Metabólica/metabolismo , Osteoblastos/metabolismo , Edulcorantes/administração & dosagem , Edulcorantes/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Western Blotting , Células da Medula Óssea/efeitos dos fármacos , Regeneração Óssea , Osso e Ossos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Dieta , Fêmur/metabolismo , Frutose/metabolismo , Masculino , Síndrome Metabólica/patologia , Osteogênese/efeitos dos fármacos , PPAR gama/metabolismo , Ratos , Ratos Sprague-Dawley , Edulcorantes/metabolismo , Fatores de Tempo , Triglicerídeos/metabolismo
10.
Diabetes Res Clin Pract ; 101(2): 177-86, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23806481

RESUMO

AIMS: Diabetes mellitus is associated with metabolic bone disease and increased low-impact fractures. The insulin-sensitizer metformin possesses in vitro, in vivo and ex vivo osteogenic effects, although this has not been adequately studied in the context of diabetes. We evaluated the effect of insulin-deficient diabetes and/or metformin on bone microarchitecture, on osteogenic potential of bone marrow progenitor cells (BMPC) and possible mechanisms involved. METHODS: Partially insulin-deficient diabetes was induced in rats by nicotinamide/streptozotocin-injection, with or without oral metformin treatment. Femoral metaphysis micro-architecture, ex vivo osteogenic potential of BMPC, and BMPC expression of Runx-2, PPARγ and receptor for advanced glycation endproducts (RAGE) were investigated. RESULTS: Histomorphometric analysis of diabetic femoral metaphysis demonstrated a slight decrease in trabecular area and a significant reduction in osteocyte density, growth plate height and TRAP (tartrate-resistant acid phosphatase) activity in the primary spongiosa. BMPC obtained from diabetic animals showed a reduction in Runx-2/PPARγ ratio and in their osteogenic potential, and an increase in RAGE expression. Metformin treatment prevented the diabetes-induced alterations in bone micro-architecture and BMPC osteogenic potential. CONCLUSION: Partially insulin-deficient diabetes induces deleterious effects on long-bone micro-architecture that are associated with a decrease in BMPC osteogenic potential, which could be mediated by a decrease in their Runx-2/PPARγ ratio and up-regulation of RAGE. These diabetes-induced alterations can be totally or partially prevented by oral administration of metformin.


Assuntos
Células da Medula Óssea/citologia , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/patologia , Insulina/deficiência , Metformina/uso terapêutico , Células-Tronco/citologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Masculino , Osteogênese/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Células-Tronco/efeitos dos fármacos
11.
Eur J Pharmacol ; 706(1-3): 41-7, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23499695

RESUMO

Accumulation of advanced glycation endproducts (AGEs) in bone tissue occurs in ageing and in Diabetes mellitus, and is partly responsible for the increased risk of low-stress bone fractures observed in these conditions. In this study we evaluated whether the anti-osteoporotic agent strontium ranelate can prevent the deleterious effects of AGEs on bone cells, and possible mechanisms of action involved. Using mouse MC3T3E1 osteoblastic cells in culture we evaluated the effects of 0.1mM strontium ranelate and/or 100 µg/ml AGEs-modified bovine serum albumin (AGEs-BSA) on cell proliferation, osteogenic differentiation and pro-inflammatory cytokine production. We found that AGEs-BSA alone decreased osteoblastic proliferation and differentiation (P<0.01) while increasing IL-1ß and TNFα production (P<0.01). On its own, strontium ranelate induced opposite effects: an increase in osteoblast proliferation and differentiation (P<0.01) and a decrease in cytokine secretion (P<0.01). Additionally, strontium ranelate prevented the inhibitory and pro-inflammatory actions of AGEs-BSA on osteoblastic cells (P<0.01). These effects of strontium ranelate were blocked by co-incubation with either the MAPK inhibitor PD98059, or the calcium channel blocker nifedipine. We also evaluated by Western blotting the activation status of ERK (a MAPK) and b-catenin. Activation of both signaling pathways was decreased by AGEs treatment, and this inhibitory effect was prevented if AGEs were co-incubated with strontium ranelate (P<0.01). On its own, strontium ranelate increased both pERK and activated b-catenin levels. In conclusion, this study demonstrates that strontium ranelate can prevent the deleterious in vitro actions of AGEs on osteoblastic cells in culture by mechanisms that involve calcium channel, MAPK and b-catenin activation.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Produtos Finais de Glicação Avançada/farmacologia , Osteoblastos/efeitos dos fármacos , Tiofenos/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Nifedipino/farmacologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , beta Catenina/metabolismo
12.
Eur J Pharmacol ; 668(3): 477-85, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21839072

RESUMO

Long-term treatment with the insulin-sensitizer rosiglitazone reduces bone mass and increases fracture risk. We have recently shown that orally administered metformin stimulates bone reossification and increases the osteogenic potential of bone marrow progenitor cells (BMPC). In the present study we investigated the effect of a 2-week metformin and/or rosiglitazone treatment on bone repair, trabecular bone microarchitecture and BMPC osteogenic potential, in young male Sprague-Dawley rats. Compared to untreated controls, rosiglitazone monotherapy decreased bone regeneration, femoral metaphysis trabecular area, osteoblastic and osteocytic density, and TRAP activity associated with epiphyseal growth plates. It also decreased the ex vivo osteogenic commitment of BMPC, inducing an increase in PPARγ expression, and a decrease in Runx2/Cbfa1 expression, in AMP-kinase phosphorylation, and in osteoblastic differentiation and mineralization. After monotherapy with metformin, with the exception of PPARγ expression which was blunted, all of the above parameters were significantly increased (compared to untreated controls). Metformin/rosiglitazone co-treatment prevented all the in vivo and ex vivo anti-osteogenic effects of rosiglitazone monotherapy, with a reversion back to control levels of PPARγ, Runx2/Cbfa1 and AMP-kinase phosphorylation of BMPC. In vitro co-incubation of BMPC with metformin and compound C-an inhibitor of AMPK phosphorylation-abrogated the metformin-induced increase in type-1 collagen production, a marker of osteoblastic differentiation. In conclusion, in rodent models metformin not only induces direct osteogenic in vivo and ex vivo actions, but when it is administered orally in combination with rosiglitazone it can prevent several of the adverse effects that this thiazolidenedione shows on bone tissue.


Assuntos
Metformina/farmacologia , Osteogênese/efeitos dos fármacos , Tiazolidinedionas/efeitos adversos , Tiazolidinedionas/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Células da Medula Óssea/citologia , Regeneração Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Colágeno Tipo I/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Interações Medicamentosas , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Fêmur/citologia , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Fêmur/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , PPAR gama/metabolismo , Ratos , Ratos Sprague-Dawley , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
13.
Acta bioquím. clín. latinoam ; 41(3): 337-346, jul.-sep. 2007. ilus, graf
Artigo em Espanhol | LILACS | ID: lil-633016

RESUMO

La hiperglucemia sostenida incrementa la glicación de proteínas. En particular, las modificaciones en las lipoproteínas de baja densidad (LDL) aumentan su potencial aterogénico. En este trabajo se comparan las modificaciones producidas por la glicación in vitro de LDL aisladas por dos métodos: precipitación selectiva (PS) y ultracentrifugación (UC). Para ello, se determinó el incremento de fructosamina, el consumo de los grupos e-amino de lisina, guanidinio de arginina y la disminución de residuos de triptofano. Para todos los analitos, los resultados cinéticos indicaron diferencias significativas con relación al basal (p<0,05), coincidentes para ambos métodos en el tiempo de aparición y en el porcentaje de variación. La aterogenicidad de las LDL glicadas separadas por PS fue estudiada en cultivos de macrófagos RAW 264.7 evaluando la formación de células espumosas y cuantificando la incorporación de LDL por tinción de los depósitos lipídicos con Oil Red. Los resultados indican que la captación de LDL modificadas aumentó con el tiempo de incubación, siendo mayor la aterogenicidad de las LDL glicadas respecto de las nativas (p<0,001, 1 h a 37 °C). El procedimiento de PS seleccionado, accesible al laboratorio bioquímico clínico, permite evaluar las modificaciones por glicación que sufren las LDL en pacientes diabéticos.


Long-term hyperglycemia increases protein glycation. In particular, modifications in the low-density lipoproteins (LDL) increase their atherogenic potential. In this study, the modifications caused by in vitro glycation of LDL separated by two methods: selective precipitation (SP) and ultracentrifugation (UC) were compared. Increase fructosamine level, decrease of e-amino group of lysine, guanidinio of arginine and triptophan fluorescence were determined. Results showed significant differences vs. basal (p<0.05) for all the tested parameters, with coincidence for the two separation methods both in time and grade of modifications. The atherogenicity of glycated LDL separated by SP was studied in macrophages RAW 264.7 in culture, through the formation of foam cells and the quantification of the dye taken up by the cellular storage lipids. Results show that the uptake of modified LDL by macrophages increased with the time of incubation, being the atherogenicity of glycated LDL greater than native LDL (p<0.001, 1 h at 37 °C). The selected SP procedure, within the facilities of routine biochemical laboratory, enables the evaluation of the modifications caused by glycation in the LDL of diabetic patients.


Assuntos
Humanos , Masculino , Feminino , Adulto , Receptor para Produtos Finais de Glicação Avançada , Lipoproteínas LDL/sangue , Apolipoproteínas , Ultracentrifugação/métodos , Técnicas de Química Analítica/métodos , Técnicas de Cultura de Células/métodos
14.
Toxicology ; 210(2-3): 205-12, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15840434

RESUMO

Bone homeostasis is the result of a tight balance between bone resorption and bone formation where macrophage activation is believed to contribute to bone resorption. We have previously shown that a vanadyl(IV)-aspirin complex (VOAspi) regulates cell proliferation and differentiation of osteoblasts in culture. In this study, we assessed VOAspi and VO effects and their possible mechanism of action on a mouse macrophage cell line RAW 264.7. Both vanadium compounds inhibited cell proliferation in a dose-dependent manner. Nifedipine completely reversed the VOAspi-induced macrophage cytotoxicity, while it could not block the effect of VO. VOAspi also stimulated nitric oxide (NO) production, the oxidation of dihydrorhodamine 123 (DHR-123) and enhanced the expression of both constitutive and inducible isoforms of nitric oxide syntases (NOS). All these effects were abolished by nifedipine. Altogether our finding give evidence that VOAspi-induced macrophage cytotoxicity is dependent on L-type calcium channel and the generation of NO though the induction of eNOS and iNOS. Contrary, the parent compound VO exerted a cytotoxic effect by mechanisms independent of a calcium entry and the NO/NOS activation.


Assuntos
Aspirina/análogos & derivados , Aspirina/toxicidade , Canais de Cálcio Tipo L/metabolismo , Proliferação de Células/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Óxido Nítrico/biossíntese , Compostos de Vanádio/toxicidade , Animais , Western Blotting , Linhagem Celular , Relação Dose-Resposta a Droga , Macrófagos/metabolismo , Camundongos , Microscopia de Fluorescência , Óxido Nítrico Sintase/biossíntese , Espécies Reativas de Oxigênio/metabolismo
15.
Acta bioquím. clín. latinoam ; 37(2): 145-151, jun. 2003. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-345609

RESUMO

El objetivo de éste estudio fue optimizar la determinación de ácido siálico en LDL aislada por precipitación selectiva y establecer relaciones entre componentes que puedan ser indicadores de aterogenicidad. Se empleó polivinilsulfato disuelto en polietilenglicol como reactivo precipitante, se ajustaron las condiciones de los lavados para garantizar la ausencia de otras proteínas del suero y se solubilizó la LDL en 5 por ciento de NaCI. Se determinó apoB, colesterol, proteínas y se optimizó la cuantificación de ácido siálico según el método de Warren modificado por Sobenin y col. Se realizó la evaluación del método analítico adaptado en cuanto a precisión intra- e inter-ensayos (CV 8 y 9 por ciento respectivamente), linealidad (hasta 7 nmol de ácido siálico/tubo); efecto de los lavados; especificidad; ausencia de interferencia de blancos de reactivo precipitante; ensayo de recuperación (entre 88 y 120 por ciento). Los resultados obtenidos sobre 30 muestras de personas normolipémicas de ambos sexos (edades entre 30 y 65 años) expresados como media ñ SEM fueron: colesterol 3,8 ñ 0,2 mmol/l, ácido siálico 34,7 ñ 2,7 µmol/l, ApoB 1,27 ñ 0,09 µmol/l, proteínas 1,57 ñ 0,08 g/l. Indices de composición: ácido siálico/colesterol: 9,24 ñ 0,48 mmol/mol, ácido siálico/apoB 34,4 ñ 3 mol/mol y ácido siálico/proteína 39,6 ñ 1,31 µmol/g. Son necesarios estudios clínicos que permitan evaluar los alcances de los índices de composición propuestos como posibles indicadores de aterogenicidad


Assuntos
Humanos , Masculino , Feminino , Ácido N-Acetilneuramínico/isolamento & purificação , Arteriosclerose , LDL-Colesterol , Técnicas de Laboratório Clínico , Lipoproteínas LDL/isolamento & purificação , Ácido N-Acetilneuramínico , Apolipoproteínas B/isolamento & purificação , Apolipoproteínas B , Precipitação Química , Colesterol , LDL-Colesterol , Lipoproteínas LDL , Fatores de Risco
16.
BMC Clin Pathol ; 1(1): 1, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11432757

RESUMO

BACKGROUND: Oxidative modification of low density lipoproteins (LDL) is recognized as one of the major processes involved in atherogenesis. The in vitro standardized measurement of LDL oxidative susceptibility could thus be of clinical significance. The aim of the present study was to establish a method which would allow the evaluation of oxidative susceptibility of LDL in the general clinical laboratory. RESULTS: LDL was isolated from human plasma by selective precipitation with amphipathic polymers. The ability of LDL to form peroxides was assessed by measuring thiobarbituric acid reactive substances (TBARS) after incubation with Cu2+ and H2O2. Reaction kinetics showed a three-phase pattern (latency, propagation and decomposition phases) which allowed us to select 150 min as the time point to stop the incubation by cooling and EDTA addition. The mixture Cu2+/H2O2 yielded more lipoperoxides than each one on its own at the same time end-point. Induced peroxidation was measured in normal subjects and in type 2 diabetic patients. In the control group, results were 21.7 +/- 1.5 nmol MDA/mg LDL protein, while in the diabetic group results were significantly increased (39.0 +/- 3.0 nmol MDA/mg LDL protein; p < 0.001). CONCLUSION: a simple and useful method is presented for the routine determination of LDL susceptibility to peroxidation in a clinical laboratory.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA