Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 425(4): 924-30, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22906741

ABSTRACT

In this study, we begin to investigate the underlying mechanism of leptin-induced vascular calcification. We found that treatment of cultured bovine aortic smooth muscle cells (BASMCs) with leptin (0.5-4 µg/ml) induced osteoblast differentiation in a dose-dependent manner. Furthermore, we found that leptin significantly increased the mRNA expression of osteopontin and bone sialoprotein, while down-regulating matrix gla protein (MGP) expression in BASMCs. Key factors implicated in osteoblast differentiation, including members of the Wnt signaling pathway, were examined. Exposure to leptin enhanced phosphorylation of GSK-3ß on serine-9 thereby inhibiting activity and promoting the nuclear accumulation of ß-catenin. Transfection of BASMCs with an adenovirus that expressed constitutively active GSK-3ß (Ad-GSK-3ß S9A) resulted in a >2-fold increase in GSK-3ß activity and a significant decrease in leptin-induced alkaline phosphatase (ALP) activity. In addition, qRT-PCR analysis showed that GSK-3ß activation resulted in a significant decrease in the expression of osteopontin and bone sialoprotein, but a marked increase in MGP mRNA expression. When taken together, our results suggest a mechanism by which leptin promotes osteoblast differentiation and vascular calcification in vivo.


Subject(s)
Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Leptin/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Osteoblasts/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Calcium-Binding Proteins/antagonists & inhibitors , Cattle , Cell Nucleus/metabolism , Cells, Cultured , Extracellular Matrix Proteins/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Integrin-Binding Sialoprotein/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Osteoblasts/cytology , Osteopontin/biosynthesis , beta Catenin/metabolism , Matrix Gla Protein
2.
J Cell Biochem ; 112(2): 581-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268079

ABSTRACT

We have previously shown that oxidized low-density lipoproteins (oxLDLs) act synergistically with ß-glycerophosphate to induce the osteogenic differentiation of primary bovine aortic smooth muscle cells (BASMCs). In the present study, we attempt to resolve the mechanism responsible for this effect by examining the expression of several osteoblast-specific transcription factors. Thus, by culturing BASMCs in the absence or presence of ß-glycerophosphate and/or oxLDL, we demonstrate that ß-glycerophosphate induces both Runx2 and Osterix (Osx) expression. In contrast, oxLDL has no effect on Runx2 expression but rather it enhances ß-glycerophosphate-induced osteoblast differentiation by further up-regulating Osx expression. In an attempt to elucidate the mechanism responsible for this latter effect, we examined the ability of oxLDL to affect Msh homeobox 2 (Msx2) expression. Similar to its effect on Osx expression, oxLDL was found to synergistically enhance ß-glycerophosphate-induced Msx2 expression in an extracellular signal-regulated kinase 1 and 2 (Erk 1 and 2)-dependent manner. Furthermore, oxLDL's ability to enhance both ß-glycerophosphate-induced Osx expression and alkaline phosphatase activity was prevented when the BASMCs were first transfected with Msx2-specific siRNA. Taken together, these findings suggest a plausible mechanism by which oxLDL may promote osteoblast differentiation and vascular calcification in vivo.


Subject(s)
Cell Differentiation/drug effects , Homeodomain Proteins/metabolism , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Transcription Factors/metabolism , Animals , Cattle , Cells, Cultured , Glycerophosphates/pharmacology , Humans , Myocytes, Smooth Muscle/cytology
3.
Arterioscler Thromb Vasc Biol ; 29(12): 2069-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19797706

ABSTRACT

OBJECTIVE: The adipocytokine leptin has been proposed to increase cardiovascular risk in both obese and diabetic individuals. In the current study, therefore, we used apoE-deficient mice to examine the effects of leptin on both lesion size and calcification. METHODS AND RESULTS: Mice were treated with once daily intraperitoneal injections of leptin (125 microg/mouse/d) for 2 months. The mice were then euthanized, and sections of the aortic root and thoracic aorta analyzed histomorphometrically. Measurements of lesion size and surface area occupied by atherosclerotic lesions did not reveal any differences between nontreated and leptin-treated animals. However, von Kossa staining of the aortic root demonstrated an 8.3+/-2.0-fold increase in lesion calcification as well as a 2.5+/-0.6-fold increase in valvular calcification in those animals treated with leptin. In addition, the percent total lesion area demonstrating ALP-positive staining was 5.4+/-2.1-fold greater in leptin-treated mice when compared to nontreated control mice. This increase in ALP staining was also accompanied by an increase in the expression of the osteoblast-specific markers, osteocalcin, and osteopontin. CONCLUSIONS: Based on these observations, we conclude that leptin may increase cardiovascular risk by promoting osteogenic differentiation and thus vascular calcification.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/etiology , Calcinosis/etiology , Leptin/pharmacology , Alkaline Phosphatase/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Valve/metabolism , Aortic Valve/pathology , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/metabolism , Atherosclerosis/pathology , Body Weight/drug effects , Calcinosis/metabolism , Calcinosis/pathology , Cattle , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Female , Humans , Lipids/blood , Mice , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/metabolism , Osteopontin/metabolism , Recombinant Proteins/pharmacology , Risk Factors
4.
Appl Physiol Nutr Metab ; 33(6): 1096-104, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19088767

ABSTRACT

Corticosteroids are a common therapy in many disease states, despite frequent and potentially serious side effects. Nutritional supplementation with conjugated linoleic acid (CLA) has been shown to increase fat-free mass, whereas supplementation with n-3 and n-6 fatty acids has been shown to increase bone mineral density (BMD). To determine whether CLA can attenuate the side effects of 8 weeks of corticosteroid administration, we randomized twenty-four 5-week-old male Sprague-Dawley rats into 1 of 4 groups: control; control + methylprednisolone (7 mg.kg-1.week-1); CLA diet (1% CLA w/w); or CLA plus methylprednisolone. Body composition, bone mineral content (BMC), and BMD were assessed with dual-energy X-ray absorptiometry at the onset and at the end of the 8-week intervention. The mechanical properties of bone were determined using 3-point femur bending at the end of the intervention. Methylprednisolone resulted in an attenuation of the increase in body mass and lean mass over the 8 weeks (p < 0.05). CLA prevented the methylprednisolone-induced attenuation of body mass and lean mass accumulation. CLA also resulted in a greater increase in BMC (p < 0.05) in the lumbar spine. The energy at failure of the isolated femurs was increased with CLA (p < 0.05). Dietary CLA prevents many of the growth- and bone-related side effects arising from 8 weeks of corticosteroid administration, results in greater increases in BMC and BMD, and can contribute to an improvement in some of the mechanical properties of bone.


Subject(s)
Bone Density/drug effects , Glucocorticoids/administration & dosage , Growth Inhibitors/administration & dosage , Growth/drug effects , Linoleic Acids, Conjugated/pharmacology , Methylprednisolone/administration & dosage , Absorptiometry, Photon/methods , Adrenal Cortex Hormones/administration & dosage , Animals , Body Composition/drug effects , Dietary Supplements , Femur/drug effects , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Time Factors
5.
Clin Exp Metastasis ; 25(8): 903-11, 2008.
Article in English | MEDLINE | ID: mdl-18814041

ABSTRACT

In the current study, we examine heparin's anti-metastatic properties by using a well-defined mouse model of osteolytic bone metastasis. C57BL/6 mice were treated with increasing doses of unfractionated heparin (15, 20, or 25 units/mouse) 30 min prior to the left ventricular injection of GFP-transfected B16F10 melanoma cells. Heparin's effect on tumour burden and bone strength was then quantified 14 days later by bone histomorphometry and biomechanical testing, respectively. Based on histomorphometric analysis of the femurs, injection of GFP-transfected melanoma cells resulted in a 37% decrease in cancellous bone volume and a 68% increase in osteoclast surface. This was associated with a 13% reduction in bone strength as measured by biomechanical testing. However, when the mice were first pre-treated with 25 units of heparin, tumour burden was decreased by 73% and tumour cell-dependent decreases in both cancellous bone volume and bone strength were prevented. Based on these observations, we conclude that heparin inhibits the ability of tumour cells to metastasize to bone and that as such, prevents tumour cell-induced decreases in bone strength.


Subject(s)
Anticoagulants/therapeutic use , Bone Neoplasms/prevention & control , Bone and Bones/drug effects , Heparin/therapeutic use , Melanoma, Experimental/prevention & control , Animals , Bone Density/physiology , Bone Neoplasms/secondary , Bone and Bones/pathology , Female , Femur/physiology , Green Fluorescent Proteins/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Osteolysis , Tumor Burden
6.
J Cell Biochem ; 105(1): 185-93, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18461557

ABSTRACT

Previous studies have localized osteoblast specific markers to sites of calcified atherosclerotic lesions. We therefore decided to use an established in vitro model of vascular calcification in order to confirm earlier reports of oxidized low-density lipoprotein (oxLDL) promoting the osteogenic differentiation of vascular smooth muscle cells. Treatment of primary bovine aortic smooth muscle cells (BASMCs) with beta-glycerophosphate was found to induce a time-dependent increase in osteoblast differentiation. In contrast, no effect was seen when BASMCs were cultured in the presence of oxLDL alone. However, when the BASMCs were cultured in the presence of both beta-glycerophosphate and oxLDL, beta-glycerophosphate's ability to induce osteoblast differentiation was significantly enhanced. In an attempt to resolve the mechanism by which this effect was occurring, we examined the effect of beta-glycerophosphate and oxLDL on several pathways known to be critical to the differentiation of osteoblasts. Surprisingly, beta-glycerophosphate alone was found to enhance Osterix (Osx) expression by inducing both Smad 1/5/8 activation and Runx2 expression. In contrast, oxLDL did not affect either Smad 1/5/8 activation or Runx2 activation but rather, it enhanced both beta-glycerophosphate-induced Osx expression and osteoblast differentiation in an extracellular signal-regulated kinase 1 and 2 (Erk 1 and 2) -dependent manner. When taken together, these findings suggest a plausible mechanism by which oxLDL may promote osteogenic differentiation and vascular calcification in vivo. J. Cell. Biochem. 105: 185-193, 2008. (c) 2008 Wiley-Liss, Inc.


Subject(s)
Cell Differentiation/drug effects , Glycerophosphates/pharmacology , Lipoproteins, LDL/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Animals , Cattle , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Drug Synergism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , RNA, Small Interfering/genetics , Smad Proteins/metabolism
7.
Thromb Res ; 122(3): 293-8, 2008.
Article in English | MEDLINE | ID: mdl-17716711

ABSTRACT

Recent clinical trials have shown that the risk of developing osteoporosis is substantially lower when low molecular weight heparins (LMWHs) are used in place of unfractionated heparin. While the reason(s) for this difference has not been fully elucidated, studies with animals have suggested that heparin causes bone loss by both decreasing bone formation and increasing bone resorption. In contrast, LMWHs appear to cause less bone loss because they only decrease bone formation. Whether all LMWHs decrease bone formation and therefore cause bone loss is unknown. For example, preliminary in vitro studies with the synthetic pentasaccaride, Fondaparinux, have suggested that it may not decrease bone formation and thus, may have no deleterious effects on bone. Further studies are required in order to determine if all LMWHs cause bone loss equally.


Subject(s)
Anticoagulants/adverse effects , Bone and Bones/drug effects , Heparin, Low-Molecular-Weight/adverse effects , Osteoporosis/chemically induced , Animals , Humans , Osteoporosis/physiopathology
8.
Med Sci Sports Exerc ; 39(5): 816-20, 2007 May.
Article in English | MEDLINE | ID: mdl-17468579

ABSTRACT

INTRODUCTION: Creatine kinase, found in osteoblasts, is an enzyme that is upregulated in response to interventions that enhance bone mass accretion. Creatine monohydrate supplementation can increase fat-free mass in young healthy men and women and can reduce markers of bone breakdown in boys with Duchenne muscular dystrophy. PURPOSE: The objective of this study was to determine the influence of supplementation with creatine monohydrate on bone structure and function in growing rats, to establish a therapeutic model. MATERIALS AND METHODS: Creatine monohydrate (2% w.w.) (CR; N = 16) or standard rat chow (CON; N = 16) was fed to Sprague-Dawley rats beginning at 5 wk of age, for 8 wk. Bone mineral density (BMD) and content (BMC) were assessed using dual-energy x-ray absorptiometry at the beginning and end of the protocol. The rats were sacrificed, and one femur was removed for the determination of mechanical properties. RESULTS: The CR-treated rats showed greater lumbar BMD and femoral bending load at failure compared with the CON rats (P < 0.05). CONCLUSIONS: Together, these data suggest that creatine monohydrate potentially has a beneficial influence on bone function and structure; further investigation is warranted into its effect on bone functional properties and its effects in disorders associated with bone loss.


Subject(s)
Bone Density/drug effects , Creatine/pharmacology , Absorptiometry, Photon , Animals , Dietary Supplements , Male , Ontario , Rats , Rats, Sprague-Dawley
9.
J Biol Chem ; 281(30): 20780-20787, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16720575

ABSTRACT

Using an animal model of heparin-induced osteoporosis we previously demonstrated that heparin causes bone loss, in part, by increasing osteoclast number and activity. Furthermore, we found that, although heparin alone has no effect, it is able to synergistically enhance Interleukin-11 (IL-11)-induced signal transducer and activator of transcription 3 (STAT3) activation and thus increase osteoclast formation in vitro. In the present study, we examine the effect of various serine kinase inhibitors on the ability of heparin to act synergistically with IL-11. Inhibition of the c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK), or the phosphatidylinositol 3-kinase pathways had no effect on the ability of heparin to promote either IL-11-induced STAT3.DNA complex formation or osteoclast formation in vitro. In contrast, PD098059, a MAPK kinase inhibitor, completely abolished the synergy between heparin and IL-11. In an attempt to resolve the mechanism by which this was occurring, we examined the effect of heparin on STAT3 Ser-727 phosphorylation and extracellular signal-regulated kinases 1 and 2 (Erk1/2) activation, either in the presence or absence of IL-11. Heparin alone was found to have no effect on Ser-727 phosphorylation, nor did heparin alter the phosphorylation status of Ser-727 in the presence of IL-11. Heparin was, however, found to increase Erk1/2 activation in both a time- and dose-dependent manner. When taken together, these findings suggest that heparin enhances IL-11-induced STAT3 activation and thus osteoclast formation, by a mechanism that is independent of STAT3 Ser-727 phosphorylation but that involves up-regulation of the MAPK pathway.


Subject(s)
Heparin/physiology , Interleukin-11/biosynthesis , MAP Kinase Signaling System , STAT3 Transcription Factor/metabolism , Signal Transduction , Up-Regulation , Animals , Cell Nucleus/metabolism , Enzyme Inhibitors/pharmacology , Female , Heparin/metabolism , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism
10.
Atherosclerosis ; 179(1): 79-86, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15721012

ABSTRACT

Recent studies have suggested that aortic smooth muscle cells undergo a phenotypic transition into osteoblast-like cells and mineralize when cultured in the presence of beta-glycerophosphate. Since we had previously demonstrated that heparin could inhibit osteoblast differentiation and mineralization in primary cultures of murine calvaria cells, we were interested in determining if heparin would have a similar effect when primary aortic smooth muscle cells were cultured in the presence of beta-glycerophosphate. The effect of heparin and low molecular weight heparin (LMWH) on osteoblast differentiation and activity was therefore examined in primary cultures of bovine aortic smooth muscle cells (BASMC) over a 14-day period. Here, we report that BASMC differentiate into osteoblast-like cells when cultured in the presence of beta-glycerophosphate. Moreover, we report that heparin not only inhibits this process but that it also inhibits the ability of BASMC to mineralize as well. Importantly, these effects were found not to be dependent upon heparins' anticoagulant activity since unfractionated heparin and heparins with low anti-thrombin III affinities inhibited the mineralization process equally well. Sulfation, however, was found to be a major determinant of heparins ability to inhibit BASMC mineralization since neither dermatan sulfate nor N-desulfated heparin were able to demonstrate an effect. We conclude that BASMC cultures can undergo a phenotypic transition into mature osteoblasts and that both the differentiation process and their ability to mineralize are inhibited by heparin.


Subject(s)
Anticoagulants/pharmacology , Heparin/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Osteoblasts/cytology , Alkaline Phosphatase/metabolism , Animals , Aorta/cytology , Calcification, Physiologic/drug effects , Cattle , Cell Differentiation/drug effects , Cells, Cultured , Glycerophosphates/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Phenotype , Sulfur/metabolism
11.
Thromb Haemost ; 92(4): 803-10, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15467912

ABSTRACT

We have previously demonstrated that heparin produces cancellous bone loss in rats due in part to a decrease in the number of osteoblasts lining the trabecular bone surface. In the present study, we use a stromal-derived cell culture system together with measurements of alkaline phosphatase (ALP) activity, to compare the effects of heparin and the low molecular weight heparin (LMWH), Fragmin, on osteoblast differentiation in vitro. In addition, we examined the possibility that both heparin and LMWH can induce adipogenesis in our stromal cell culture system. Both heparin and LMWH were found to produce a statistically significant (P < 0.01) and concentration-dependent decrease in the number of osteoblasts while increasing the number of adipocytes. When the effects of gravimetrically equivalent amounts of heparin and LMWH were compared, heparin had a 4-fold greater effect than LMWH. In contrast to heparin, N-desulfated heparin was found to have minimal effects on both osteoblast and adipocyte differentiation indicating that the heparin effect is not only chain-length dependent but also charge-dependent. The observation that LMWH has less of an effect on bone formation than heparin is compatible with the results of clinical trials indicating that LMWH produces less bone loss after long-term administration.


Subject(s)
Adipocytes/cytology , Cell Differentiation/drug effects , Heparin, Low-Molecular-Weight/pharmacology , Heparin/pharmacology , Osteoblasts/cytology , Adipocytes/drug effects , Animals , Biomarkers/analysis , Cell Count , Femur/cytology , Mice , Osteoblasts/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects
12.
Thromb Haemost ; 91(5): 927-34, 2004 May.
Article in English | MEDLINE | ID: mdl-15116253

ABSTRACT

Unfractionated heparin (UFH) and low-molecular-weight heparin (LMWH) are cleared, at least in part, by the kidneys through a poorly understood process. This study was undertaken to explore the mechanism of renal clearance of these drugs. Rats were given fluorescein-5-isothiocyanate (FITC)-labeled UFH or LMWH intravenously. At intervals after injection, rats were euthanized and the kidneys were harvested and subjected to immunohistochemical analysis and fluorescence microscopy. Both UFH and LMWH were localized to renal tubular cells and no immunoperoxidase staining or fluorescence was detected in glomeruli. Autoradiography demonstrated similar intracellular distribution of radio-labeled UFH suggesting that this phenomenon is independent of the method used to label heparin. Fluorescence in the tubules increased as a function of time after UFH injection, but reached a plateau after LMWH injection suggesting that the rate of renal tubular uptake depends on the molecular size of the heparin. When administered prior to FITC-labeled UFH or LMWH, probenecid, a renal organic anion inhibitor, decreased the renal tubular uptake of the heparins, whereas cimetidine, a renal organic cation inhibitor, had no effect. These findings suggest that renal excretion of UFH and LMWH primarily reflects tubular uptake via an organic anion transport mechanism.


Subject(s)
Heparin, Low-Molecular-Weight/pharmacokinetics , Heparin/pharmacokinetics , Kidney/metabolism , Animals , Anions/pharmacology , Drug Evaluation, Preclinical , Fluorescein-5-isothiocyanate , Immunohistochemistry , Kidney Tubules/cytology , Kidney Tubules/metabolism , Kinetics , Male , Molecular Weight , Probenecid/pharmacology , Rats , Rats, Sprague-Dawley , Tissue Distribution
13.
Cytokine ; 20(2): 78-85, 2002 Oct 21.
Article in English | MEDLINE | ID: mdl-12445802

ABSTRACT

The issue of whether interleukin-11 (IL-11) contributes to bone loss during states of estrogen deficiency has not been previously determined. We therefore randomized ovariectomized (OVX) mice to once daily interperitoneal injections of either sheep anti-murine IL-11 Ab or normal sheep IgG (NSIgG) for 21 days, and then determined the effects on bone using bone histomorphometry. Here we report that treatment of OVX mice with anti-IL-11 Ab significantly increases both trabecular width and cancellous bone volume. Osteoblast activity, as measured by the percentage of trabecular surface covered by osteoid and rates of bone formation, were also significantly increased following treatment with anti-IL-11 Ab. In contrast, treatment of OVX mice with anti-IL-11 Ab significantly decreased both osteoclast number and activity. Ex-vivo assays of osteoclast formation and activity confirmed the histomorphometric data. Thus, bone marrow cells isolated from anti-IL-11 Ab treated OVX mice formed fewer osteoclasts and resorbed less bone in culture than did marrow cells isolated from either untreated or NSIgG-treated OVX mice. Based on these results we conclude that IL-11 contributes to the bone loss which is observed during states of estrogen deficiency.


Subject(s)
Bone Remodeling/immunology , Interleukin-11/antagonists & inhibitors , Osteoclasts/cytology , Osteoclasts/immunology , Animals , Bone Remodeling/physiology , Estrogens/deficiency , Female , In Vitro Techniques , Interleukin-11/physiology , Mice , Mice, Inbred C3H , Neutralization Tests , Ovariectomy , Sheep
14.
Blood ; 100(7): 2530-6, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12239166

ABSTRACT

We have previously demonstrated that long-term heparin treatment causes cancellous bone loss in rats due in part to an increase in the number of osteoclasts lining the trabecular bone surface. In the present study, we investigated this phenomenon by examining the ability of heparin to synergistically enhance interleukin-11 (IL-11)-induced osteoclast formation. Treatment of murine calvaria and bone marrow cells with IL-11 was found to induce the formation of tartrate-resistant acid phosphatase-positive (TRAP(+)) multinucleated cells (MNCs) in a dose-dependent fashion. No effect was seen when cocultures were treated with heparin alone. However, when cocultures were treated with both IL-11 and heparin, IL-11's ability to induce TRAP(+) MNC formation was enhanced 6-fold. In an attempt to resolve the mechanism responsible for this effect, we examined the ability of heparin to influence IL-11 signaling using murine calvaria cells. Heparin was found to enhance both IL-11-induced STAT3-DNA complex formation and transactivation without altering either STAT3 (signal transducer and activator of transcription-3) tyrosine or serine phosphorylation. Heparin was also found to enhance IL-11's ability to induce the expression of both receptor activator of nuclear factor-kappaB ligand (RANKL) and glycoprotein (gp) 130. When taken together, these findings suggest a plausible mechanism by which heparin may cause increased osteoclastogenesis and therefore bone loss when administered long-term.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , DNA-Binding Proteins/metabolism , Heparin/pharmacology , Interleukin-11/pharmacology , Osteoclasts/physiology , Skull/cytology , Trans-Activators/metabolism , Acute-Phase Proteins/metabolism , Animals , Animals, Newborn , Blotting, Northern , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Drug Synergism , Female , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Osteoclasts/drug effects , Phosphates/metabolism , Phosphorylation , Pregnancy , STAT3 Transcription Factor , Skull/drug effects , Skull/physiology
15.
Thromb Res ; 105(4): 353-8, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-12031831

ABSTRACT

The issue of whether long-term sodium warfarin therapy results in decreased bone density is controversial. To address this question, we randomized rats to once daily subcutaneous injections of either sodium warfarin (0.20 or 0.25 mg/kg) or saline for 28 days and monitored the effects on bone, both biomechanically and by histomorphometric analysis. In addition, the anticoagulant status of both saline- and warfarin-treated rats were monitored throughout the course of the experiment by measuring the prothrombin time, expressed as international normalized ratios (INRs). Rats treated with 0.25 mg/kg warfarin demonstrated INRs of approximately 2.6, while rats treated with either 0.20 mg/kg warfarin or saline were found to have INRs of 1.3 and 1.0, respectively. Biomechanical testing of the right femur of rats treated with 0.25 mg/kg warfarin demonstrated that warfarin caused an 8% reduction in bone strength as measured by maximum tolerated load. A similar reduction in the biomechanical parameters of energy to break (P<.0001) and force at break point (P<.005) was also observed. Histomorphometric analysis of the left femur of warfarin-treated rats revealed a 17% reduction in cancellous bone volume. This was accompanied by a 60% decrease in osteoblast surface, as well as an 80% reduction in osteoid surface. In contrast, warfarin treatment had the opposite effect on osteoclast surface, which was 35% higher following warfarin treatment. Based on these observations, we conclude that clinically relevant doses of warfarin decrease femoral bone strength and cancellous bone volume, both by decreasing the rate of bone formation and increasing the rate of bone resorption.


Subject(s)
Anticoagulants/toxicity , Femur/drug effects , Femur/pathology , Warfarin/toxicity , Animals , Biomechanical Phenomena , Bone Density/drug effects , Bone Remodeling/drug effects , Femur/physiopathology , Humans , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoporosis/chemically induced , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL