Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Bone ; 94: 141-151, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27989651

ABSTRACT

Skeletal co-morbidities in type 1 diabetes include an increased risk for fracture and delayed fracture healing, which are intertwined with disease duration and the presence of other diabetic complications. As such, chronic hyperglycemia is undoubtedly a major contributor to these outcomes, despite standard insulin-replacement therapy. Therefore, using the streptozotocin (STZ)-induced model of hypoinsulinemic hyperglycemia in DBA/2J male mice, we compared the effects of two glucose lowering therapies on the fracture resistance of bone and markers of bone turnover. Twelve week-old diabetic (DM) mice were treated for 9weeks with: 1) oral canagliflozin (CANA, dose range ~10-16mg/kg/day), an inhibitor of the renal sodium-dependent glucose co-transporter type 2 (SGLT2); 2) subcutaneous insulin, via minipump (INS, 0.125units/day); 3) co-therapy (CANA+INS); or 4) no treatment (STZ, without therapy). These groups were also compared to non-diabetic control groups. Untreated diabetic mice experienced increased bone resorption and significant deficits in cortical and trabecular bone that contributed to structural weakness of the femur mid-shaft and the lumbar vertebra, as determined by three-point bending and compression tests, respectively. Treatment with either canagliflozin or insulin alone only partially rectified hyperglycemia and the diabetic bone phenotype. However, when used in combination, normalization of glycemic control was achieved, and a prevention of the DM-related deterioration in bone microarchitecture and bone strength occurred, due to additive effects of canagliflozin and insulin. Nevertheless, CANA-treated mice, whether diabetic or non-diabetic, demonstrated an increase in urinary calcium loss; FGF23 was also increased in CANA-treated DM mice. These findings could herald ongoing bone mineral losses following CANA exposure, suggesting that certain CANA-induced skeletal consequences might detract from therapeutic improvements in glycemic control, as they relate to diabetic bone disease.


Subject(s)
Bone Diseases, Metabolic/drug therapy , Canagliflozin/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Insulin/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors , Animals , Biomarkers/metabolism , Blood Glucose/metabolism , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/complications , Bone Resorption/blood , Bone Resorption/complications , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/pathology , Canagliflozin/pharmacology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Disease Models, Animal , Fibroblast Growth Factor-23 , Insulin/pharmacology , Linear Models , Male , Mice, Inbred DBA , Phenotype , Sodium-Glucose Transporter 2/metabolism
3.
J Orthop Res ; 34(10): 1716-1724, 2016 10.
Article in English | MEDLINE | ID: mdl-26867804

ABSTRACT

The majority of Osteosarcoma (OS) patients are treated with a combination of chemotherapy, resection, and limb salvage protocols. These protocols include distraction osteogenesis (DO), which is characterized by direct new bone formation. Cisplatin (CDP) is extensively used for OS chemotherapy and recent studies, using a mouse DO model, have demonstrated that CDP has profound negative effects on bone repair. Recent oncological therapeutic strategies are based on the use of standard cytotoxic drugs plus an assortment of biologic agents. Here we demonstrate that the previously reported CDP-associated inhibition of bone repair can be modulated by the administration of a small molecule p53 inducer (nutlin-3). The effects of nutlin-3 on CDP osteotoxicity were studied using both pre- and post-operative treatment models. In both cases the addition of nutlin-3, bracketing CDP exposure, demonstrated robust and significant bone sparing activity (p < 0.01-0.001). In addition the combination of nutlin-3 and CDP induced equivalent OS tumor killing in a xenograft model. Collectively, these results demonstrate that the induction of p53 peri-operatively protects bone healing from the toxic effects of CDP, while maintaining OS toxicity. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1716-1724, 2016.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Regeneration/drug effects , Cisplatin/therapeutic use , Imidazoles/therapeutic use , Osteosarcoma/drug therapy , Piperazines/therapeutic use , Animals , Female , Humans , Imidazoles/pharmacology , Male , Mice, Inbred C57BL , Mice, Nude , Osteogenesis, Distraction , Osteosarcoma/surgery , Piperazines/pharmacology , Random Allocation , Xenograft Model Antitumor Assays
4.
Bone ; 82: 101-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26211996

ABSTRACT

Persons with type 1 and type 2 diabetes have increased fracture risk, attributed to deficits in the microarchitecture and strength of diabetic bone, thought to be mediated, in part, by the consequences of chronic hyperglycemia. Therefore, to examine the effects of a glucose-lowering SGLT2 inhibitor on blood glucose (BG) and bone homeostasis in a model of diabetic bone disease, male DBA/2J mice with or without streptozotocin (STZ)-induced hyperglycemia were fed chow containing the SGLT2 inhibitor, canagliflozin (CANA), or chow without drug, for 10weeks of therapy. Thereafter, serum bone biomarkers were measured, fracture resistance of cortical bone was assessed by µCT analysis and a three-point bending test of the femur, and vertebral bone strength was determined by compression testing. In the femur metaphysis and L6 vertebra, long-term diabetes (DM) induced deficits in trabecular bone microarchitecture. In the femur diaphysis, a decrease in cortical bone area, cortical thickness and minimal moment of inertia occurred in DM (p<0.0001, for all) while cortical porosity was increased (p<0.0001). These DM changes were associated with reduced fracture resistance (decreased material strength and toughness; decreased structural strength and rigidity; p<0.001 for all). Significant increases in PTH (p<0.0001), RatLAPs (p=0.0002), and urine calcium concentration (p<0.0001) were also seen in DM. Canagliflozin treatment improved BG in DM mice by ~35%, but did not improve microarchitectural parameters. Instead, in canagliflozin-treated diabetic mice, a further increase in RatLAPs was evident, possibly suggesting a drug-related intensification of bone resorption. Additionally, detrimental metaphyseal changes were noted in canagliflozin-treated control mice. Hence, diabetic bone disease was not favorably affected by canagliflozin treatment, perhaps due to insufficient glycemic improvement. Instead, in control mice, long-term exposure to SGLT2 inhibition was associated with adverse effects on the trabecular compartment of bone.


Subject(s)
Blood Glucose/drug effects , Bone Diseases/drug therapy , Canagliflozin/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Sodium-Glucose Transporter 2 Inhibitors , Animals , Blood Glucose/metabolism , Bone Diseases/metabolism , Bone Diseases/prevention & control , Canagliflozin/pharmacology , Diabetes Mellitus, Experimental/metabolism , Male , Mice , Mice, Inbred DBA , Sodium-Glucose Transporter 2/metabolism
5.
Bone Rep ; 1: 16-19, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25685827

ABSTRACT

In type 1 diabetes, diabetic bone disease (DBD) is characterized by decreased bone mineral density, a state of low bone turnover and an increased risk of fracture. Animal models of DBD demonstrate that acquired alterations in trabecular and cortical bone microarchitecture contribute to decreased bone strength in diabetes. With anti-collagenolytic and anti-inflammatory properties, tetracycline derivatives may prevent diabetes-related decreases in bone strength. To determine if doxycycline, a tetracycline class antibiotic, can prevent the development of DBD in a model of long-term diabetes, male DBA/2J mice, with or without diabetes, were treated with doxycycline-containing chow for 10 weeks (dose range, 28-92 mg/kg/day). Long-term doxycycline exposure was not deleterious to the microarchitecture or biomechanical properties of healthy bone in male DBA/2J mice. Doxycycline treatment also did not prevent or alleviate the deleterious changes in trabecular microarchitecture, cortical structure, and biomechanical properties of bone induced by chronic diabetes.

6.
J Diabetes Res ; 2014: 703589, 2014.
Article in English | MEDLINE | ID: mdl-24963495

ABSTRACT

Type 1 diabetes mellitus (T1D) is associated with decreased bone mineral density, a deficit in bone structure, and subsequently an increased risk of fragility fracture. These clinical observations, paralleled by animal models of T1D, suggest that the insulinopenia of T1D has a deleterious effect on bone. To further examine the action of insulin signaling on bone development, we generated mice with an osteoprogenitor-selective (osterix-Cre) ablation of the insulin receptor (IR), designated OIRKO. OIRKO mice exhibited an 80% decrease in IR in osteoblasts. Prenatal elimination of IR did not affect fetal survival or gross morphology. However, loss of IR in mouse osteoblasts resulted in a postnatal growth-constricted phenotype. By 10-12 weeks of age, femurs of OIRKO mice were more slender, with a thinner diaphyseal cortex and, consequently, a decrease in whole bone strength when subjected to bending. In male mice alone, decreased metaphyseal trabecular bone, with thinner and more rodlike trabeculae, was also observed. OIRKO mice did not, however, exhibit abnormal glucose tolerance. The skeletal phenotype of the OIRKO mouse appeared more severe than that of previously reported bone-specific IR knockdown models, and confirms that insulin receptor expression in osteoblasts is critically important for proper bone development and maintenance of structural integrity.


Subject(s)
Bone and Bones/pathology , Osteoblasts/cytology , Receptor, Insulin/physiology , Animals , Biomechanical Phenomena , Body Size , Female , Genotype , Glucose/metabolism , Insulin/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Osteoblasts/metabolism , Osteogenesis , Phenotype , Receptor, Insulin/genetics
7.
Alcohol Clin Exp Res ; 38(3): 672-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24256560

ABSTRACT

BACKGROUND: In bone, NADPH oxidase (NOX)-derived reactive oxygen species (ROS) superoxide and/or hydrogen peroxide are an important stimulus for osteoclast differentiation and activity. Previously, we have demonstrated that chronic ethanol (EtOH) consumption generates excess NOX-dependent ROS in osteoblasts, which functions to stimulate nuclear factor kappa-ß receptor ligand (RANKL)-RANK signaling, thus increasing osteoclastogenesis and activity. This activity can be blocked by co-administration of EtOH with the pan-NOX inhibitor diphenylene idonium (DPI). METHODS: To test whether EtOH-induced bone loss is dependent on a functional NOX2 enzyme, 6-week-old female C57BL/6J-Ncf1/p47phox(-/-) (p47phox KO) and wild-type (WT) mice were pair-fed EtOH diets for 40 days. Bone loss was assessed by 3-point bending, micro-computed tomography and static histomorphometric analysis. Additionally, ST2 cultured cells were co-treated with EtOH and NOX inhibitors, DPI, gliotoxin, and plumbagin, after which changes in ROS production, and in RANKL and NOX mRNA expression were analyzed. RESULTS: In WT mice, EtOH treatment significantly reduced bone density and mechanical strength, and increased total osteoclast number and activity. In EtOH-treated p47phox KO mice, bone density and mechanical strength were completely preserved. EtOH p47phox KO mice had no changes in osteoclast numbers or activity, and no elevations in serum CTX or RANKL gene expression (p < 0.05). In both WT and p47phox KO mice, EtOH feeding reduced biochemical markers of bone formation (p < 0.05). In vitro EtOH exposure of ST2 cells increased ROS, which was blocked by pretreating with DPI or the NOX2 inhibitor gliotoxin. EtOH-induced RANKL and NOX2 gene expression were inhibited by the NOX4-specific inhibitor plumbagin. CONCLUSIONS: These data suggest that NOX2-derived ROS is necessary for EtOH-induced bone resorption. In osteoblasts, NOX2 and NOX4 appear to work in tandem to increase RANKL expression, whereas EtOH-mediated inhibition of bone formation occurs via a NOX2-independent mechanism.


Subject(s)
Bone Resorption/chemically induced , Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Bone Resorption/enzymology , Cells, Cultured , Female , Genotype , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Osteogenesis/drug effects , RANK Ligand/metabolism , Random Allocation
8.
J Orthop Res ; 32(3): 464-70, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24259375

ABSTRACT

Osteosarcoma (OS) is the most common malignant bone tumor affecting children and adolescents. Many patients are treated with a combination of chemotherapy, resection, and limb salvage protocols. Surgical reconstructions after tumor resection include structural allografts, non-cemented endoprostheses, and distraction osteogenesis (DO), which require direct bone formation. Although cisplatin (CDP) is extensively used for OS chemotherapy, the effects on bone regeneration are not well studied. The effects of CDP on direct bone formation in DO were compared using two dosing regimens and both C57BL/6 (B6) and tumor necrosis factor receptor 1 knockout (TNFR1KO) mice, as CDP toxicity is associated with elevated TNF levels. Detailed evaluation of the five-dose CDP regimen (2 mg/kg/day), demonstrated significant decreases in new bone formation in the DO gaps of CDP treated versus vehicle treated mice (p < 0.001). Further, no significant inhibitory effects from the five-dose CDP regimen were observed in TNFR1KO mice. The two-dose regimen significantly inhibited new bone formation in B6 mice. These results demonstrate that CDP has profound short term negative effects on the process of bone repair in DO. These data provide the mechanistic basis for modeling peri-operative chemotherapy doses and schedules and may provide new opportunities to identify molecules that spare normal cells from the inhibitory effects of CDP.


Subject(s)
Antineoplastic Agents/toxicity , Bone Regeneration/drug effects , Cisplatin/toxicity , Osteogenesis, Distraction , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor-alpha/metabolism
9.
Bone ; 57(1): 36-40, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23886838

ABSTRACT

OBJECTIVE: Using a streptozotocin (STZ)-induced mouse model of type 1 diabetes (T1D), we have previously demonstrated that long-term diabetes inhibits regenerative bone formation during tibial distraction osteogenesis (DO) and perturbs skeletal integrity by decreasing cortical thickness, bone mineral density and bone's resistance to fracture. Because long-standing T1D is also associated with a deficiency of insulin-like growth factor I (IGF-I), we examined the effects of systemic IGF-I treatment on skeletal microarchitecture and strength, as well as on bone formation in diabetic mice. RESEARCH DESIGN AND METHODS: Streptozotocin-induced diabetic or control mice were treated with recombinant human IGF-I (rhIGF-I, 1.5mg/kg/day as subcutaneous infusion) or vehicle throughout a 14day DO procedure. Thereafter, trunk blood was assayed for glucose, insulin, rhIGF-I, mouse IGF-I and leptin. Bone formation in distracted tibiae was quantified. Effects on cortical bone strength and trabecular bone architecture were assessed by µCT analysis and three-point bend testing of contralateral femurs. RESULTS: New bone formation during DO was reduced in diabetic mice but significantly improved with rhIGF-I treatment. The contralateral femurs of diabetic mice demonstrated significant reductions in trabecular thickness, yield strength and peak force of cortical bone, which were improved with rhIGF-I treatment. rhIGF-I also reduced intracortical porosity in control mice. However, treatment with rhIGF-I did not normalize serum glucose, or correct concurrent deficiencies of insulin or leptin seen in diabetes. CONCLUSIONS: These findings demonstrate that despite persistent hyperglycemia, rhIGF-I promoted new bone formation and improved biomechanical properties of bone in a model of T1D, suggesting that it may be useful as a fracture preventative in this disease.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Insulin-Like Growth Factor I/therapeutic use , Animals , Hyperglycemia/drug therapy , Mice , Osteogenesis/drug effects
10.
J Pharmacol Exp Ther ; 343(2): 401-12, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22892342

ABSTRACT

Chronic alcohol abuse results in decreased bone mineral density (BMD), which can lead to increased fracture risk. In contrast, low levels of alcohol have been associated with increased BMD in epidemiological studies. Alcohol's toxic skeletal effects have been suggested to involve impaired vitamin D/calcium homeostasis. Therefore, dietary vitamin D supplementation may be beneficial in reducing bone loss associated with chronic alcohol consumption. Six-week-old female C57BL/6J mice were pair-fed ethanol (EtOH)-containing liquid diets (10 or 36% total calories) for 78 days. EtOH exposure at 10% calories had no effects on any measured bone or serum parameter. EtOH consumption at 36% of calories reduced BMD and bone strength (P<0.05), decreased osteoblastogenesis, increased osteoclastogenesis, suppressed 1,25-hydroxyvitamin D3 [1,25(OH)2D3] serum concentrations (P<0.05), and increased apoptosis in bone cells compared with pair-fed controls. In a second study, female mice were pair-fed 30% EtOH diets with or without dietary supplementation with vitamin D3 (cholecalciferol; VitD) for 40 days. VitD supplementation in the EtOH diet protected against cortical bone loss, normalized alcohol-induced hypocalcaemia, and suppressed EtOH-induced expression of receptor of nuclear factor-κB ligand mRNA in bone. In vitro, pretreatment of 1,25(OH)2D3 in osteoblastic cells inhibited EtOH-induced apoptosis. In EtOH/VitD mice circulating 1,25(OH)2D3 was lower compared with mice receiving EtOH alone (P<0.05), suggesting increased sensitivity to feedback control of VitD metabolism in the kidney. These findings suggest dietary VitD supplementation may prevent skeletal toxicity in chronic drinkers by normalizing calcium homeostasis, preventing apoptosis, and suppressing EtOH-induced increases in bone resorption.


Subject(s)
Bone Density/drug effects , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Osteoporosis, Postmenopausal/prevention & control , Vitamin D/pharmacology , Vitamins/pharmacology , Animals , Apoptosis/drug effects , Biomechanical Phenomena , Body Composition/drug effects , Body Weight/drug effects , Bone Marrow/drug effects , Bone Remodeling/drug effects , Cells, Cultured , Central Nervous System Depressants/antagonists & inhibitors , Cholecalciferol/blood , Cholecalciferol/pharmacology , Ethanol/antagonists & inhibitors , Female , Femur/pathology , Gene Expression/drug effects , Humans , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/chemically induced , RNA/biosynthesis , RNA/genetics , Tomography, X-Ray Computed , Vitamin D/blood , Vitamins/blood , Weight Gain/drug effects
11.
Alcohol ; 46(2): 133-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21908154

ABSTRACT

Distraction osteogenesis (DO) is an orthopedic protocol, which induces direct new bone formation as a result of the stimulating effects of mechanical distraction. Chronic ethanol exposure has been demonstrated to inhibit bone formation in rodent models of DO. Further, it has been demonstrated that (1) tumor necrosis factor-α (TNF) blockers are protective against ethanol exposure and (2) recombinant mouse TNF (rmTNF) inhibits direct bone formation in ethanol naïve mice through TNF receptor 1 (TNFR1). These results suggest that the inhibitory effects are significantly mediated by TNF signaling. Therefore, we hypothesized that direct new bone formation in TNFR1 knockout (KO) mice would be protected from ethanol exposure. We used a unique model of mouse DO combined with liquid/chow diets to compare the effects of ethanol on both a strain of TNFR1 knockout (TNFR1 KO) mice and on mice of their C57BL/6 (B6) control strain. In the B6 study, and in concordance with previous work, both radiological and histological analyses of direct bone formation in the distraction gaps demonstrated significant osteoinhibition due to ethanol compared with chow- or pair-fed mice. In the TNFR1 KO study and in support of the hypothesis, both radiological and histological analyses of distraction gap bone formation demonstrated no significant differences between the ethanol, chow fed, or pair fed. We conclude that exogenous rmTNF and ethanol-induced endogenous TNF act to inhibit new bone formation during DO by signaling primarily through TNFR1.


Subject(s)
Ethanol/pharmacology , Osteogenesis, Distraction/methods , Osteogenesis/drug effects , Tumor Necrosis Factor-alpha/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/deficiency , Signal Transduction/drug effects , Tibia/drug effects , Tibia/growth & development , Tibial Fractures/surgery
12.
Exp Diabetes Res ; 2011: 269378, 2011.
Article in English | MEDLINE | ID: mdl-21747824

ABSTRACT

Microalbuminuria in humans with Type 1 diabetes (T1D) is associated with increased urinary excretion of megalin, as well as many megalin ligands, including vitamin-D-binding protein (VDBP). We examined the DBA/2J diabetic mouse, nephropathy prone model, to determine if megalin and VDBP excretion coincide with the development of diabetic nephropathy. Megalin, VDBP, and 25-hydroxy-vitamin D (25-OHD) were measured in urine, and genes involved in vitamin D metabolism were assessed in renal tissues from diabetic and control mice at 10, 15, and 18 weeks following the onset of diabetes. Megalin, VDBP, and 25-OHD were increased in the urine of diabetic mice. 1-α hydroxylase (CYP27B1) mRNA in the kidney was persistently increased in diabetic mice, as were several vitamin D-target genes. These studies show that intrarenal vitamin D handling is altered in the diabetic kidney, and they suggest that in T1D, urinary losses of VDBP may portend risk for intrarenal and extrarenal vitamin D deficiencies.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Endocytosis , Vitamin D/metabolism , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/genetics , Diabetic Nephropathies/pathology , Disease Models, Animal , Disease Susceptibility , Endocytosis/physiology , Kidney/metabolism , Kidney/pathology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice , Mice, Inbred DBA , Signal Transduction/physiology , Streptozocin
13.
J Pharmacol Exp Ther ; 336(3): 734-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21098090

ABSTRACT

Previous in vitro data suggest that ethanol (EtOH) activates NADPH oxidase (Nox) in osteoblasts leading to accumulation of reactive oxygen species (ROS). This might be a mechanism underlying inhibition of bone formation and increased bone resorption observed in vivo after EtOH exposure. In a rat model in which cycling females were infused intragastrically with EtOH-containing liquid diets, EtOH significantly decreased bone formation and stimulated osteoblast-dependent osteoclast differentiation. These effects were reversed by exogenous 17-ß-estradiol coadministration. Moreover, coadministration of N-acetyl cysteine (NAC), an antioxidant, or diphenylene iodonium (DPI), a specific Nox inhibitor, also abolished chronic EtOH-associated bone loss. EtOH treatment up-regulated mRNA levels of Nox1, 2, 4, and the receptor activator of nuclear factor-κB ligand (RANKL), an essential factor for differentiation of osteoclasts in bone. Protein levels of Nox4, a major Nox isoform expressed in nonphagocytic cells, was also up-regulated by EtOH in bone. 17-ß-Estradiol, NAC, and DPI were able to normalize EtOH-induced up-regulation of Nox and RANKL. In vitro experiments demonstrated that EtOH directly up-regulated Nox expression in osteoblasts. Pretreatment of osteoblasts with DPI eliminated EtOH-induced RANKL promoter activity. Furthermore, EtOH induced RANKL gene expression, and RANKL promoter activation in osteoblasts was ROS-dependent. These data suggest that inhibition of Nox expression and activity may be critical for prevention of chronic EtOH-induced osteoblast-dependent bone loss.


Subject(s)
Bone Resorption/enzymology , Bone Resorption/prevention & control , Ethanol/toxicity , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Animals , Bone Resorption/chemically induced , Cell Line , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Onium Compounds/pharmacology , Onium Compounds/therapeutic use , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
14.
Bone ; 48(4): 733-40, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21185416

ABSTRACT

Type 1 diabetes (T1DM) increases the likelihood of a fracture. Despite serious complications in the healing of fractures among those with diabetes, the underlying causes are not delineated for the effect of diabetes on the fracture resistance of bone. Therefore, in a mouse model of T1DM, we have investigated the possibility that a prolonged state of diabetes perturbs the relationship between bone strength and structure (i.e., affects tissue properties). At 10, 15, and 18 weeks following injection of streptozotocin to induce diabetes, diabetic male mice and age-matched controls were examined for measures of skeletal integrity. We assessed 1) the moment of inertia (I(MIN)) of the cortical bone within diaphysis, trabecular bone architecture of the metaphysis, and mineralization density of the tissue (TMD) for each compartment of the femur by micro-computed tomography and 2) biomechanical properties by three-point bending test (femur) and nanoindentation (tibia). In the metaphysis, a significant decrease in trabecular bone volume fraction and trabecular TMD was apparent after 10 weeks of diabetes. For cortical bone, type 1 diabetes was associated with decreased cortical TMD, I(MIN), rigidity, and peak moment as well as a lack of normal age-related increases in the biomechanical properties. However, there were only modest differences in material properties between diabetic and normal mice at both whole bone and tissue-levels. As the duration of diabetes increased, bone toughness decreased relative to control. If the sole effect of diabetes on bone strength was due to a reduction in bone size, then I(MIN) would be the only significant variable explaining the variance in the maximum moment. However, general linear modeling found that the relationship between peak moment and I(MIN) depended on whether the bone was from a diabetic mouse and the duration of diabetes. Thus, these findings suggest that the elevated fracture risk among diabetics is impacted by complex changes in tissue properties that ultimately reduce the fracture resistance of bone.


Subject(s)
Bone and Bones/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Animals , Bone Density , Diabetes Mellitus, Experimental/chemically induced , Male , Mice , Mice, Inbred DBA , Streptozocin , Tomography, X-Ray Computed
15.
Cardiovasc Diabetol ; 9: 73, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-21054880

ABSTRACT

BACKGROUND: Insulin resistance is associated with a proinflammatory state that promotes the development of complications such as type 2 diabetes mellitus (T2DM) and atherosclerosis. The metabolic stimuli that initiate and propagate proinflammatory cytokine production and the cellular origin of proinflammatory cytokines in insulin resistance have not been fully elucidated. Circulating proinflammatory monocytes show signs of enhanced inflammation in obese, insulin resistant subjects and are thus a potential source of proinflammatory cytokine production. The specific, circulating metabolic factors that might stimulate monocyte inflammation in insulin resistant subjects are poorly characterized. We have examined whether saturated nonesterified fatty acids (NEFA) and insulin, which increase in concentration with developing insulin resistance, can trigger the production of interleukin (IL)-6 and tumor necrosis factor (TNF)-α in human monocytes. METHODS: Messenger RNA and protein levels of the proinflammatory cytokines IL-6 and TNF-α were measured by quantitative real-time PCR (qRT-PCR) and Luminex bioassays. Student's t-test was used with a significance level of p < 0.05 to determine significance between treatment groups. RESULTS: Esterification of palmitate with coenzyme A (CoA) was necessary, while ß-oxidation and ceramide biosynthesis were not required, for the induction of IL-6 and TNF-α in THP-1 monocytes. Monocytes incubated with insulin and palmitate together produced more IL-6 mRNA and protein, and more TNF-α protein, compared to monocytes incubated with palmitate alone. Incubation of monocytes with insulin alone did not affect the production of IL-6 or TNF-α. Both PI3K-Akt and MEK/ERK signalling pathways are important for cytokine induction by palmitate. MEK/ERK signalling is necessary for synergistic induction of IL-6 by palmitate and insulin. CONCLUSIONS: High levels of saturated NEFA, such as palmitate, when combined with hyperinsulinemia, may activate human monocytes to produce proinflammatory cytokines and support the development and propagation of the subacute, chronic inflammatory state that is characteristic of insulin resistance. Results with inhibitors of ß-oxidation and ceramide biosynthesis pathways suggest that increased fatty acid flux through the glycerolipid biosynthesis pathway may be involved in promoting proinflammatory cytokine production in monocytes.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Inflammation Mediators/metabolism , Insulin/metabolism , Interleukin-6/metabolism , Monocytes/metabolism , Palmitic Acid/metabolism , Cell Line, Tumor , Ceramides/biosynthesis , Coenzyme A/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Insulin Resistance , Interleukin-6/genetics , MAP Kinase Kinase Kinases/metabolism , Monocytes/immunology , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
16.
Endocrine ; 37(2): 336-43, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20960272

ABSTRACT

Neutrophil gelatinase-associated lipocalin (NGAL), a biomarker of renal injury, can bind matrix metalloproteinase-9 (MMP-9) and inhibit its degradation, thereby sustaining MMP-9 proteolytic activity. MMP-9 is produced by renal podocytes, and podocyte MMP production can be modified by high ambient glucose levels. Moreover, dysregulation of MMP-9 activity, gene expression, or urine concentrations has been demonstrated in T2DM-associated nephropathy and in non-diabetic proteinuric renal diseases. Our objective was to determine whether NGAL/MMP-9 dysregulation might contribute to or serve as a biomarker of diabetic nephropathy in type 1 DM (T1DM). Plasma MMP-9, and urine NGAL and MMP-9 concentrations were measured in 121 T1DM and 55 control subjects and examined relative to indicators of glycemia, renal function, and degree of albuminuria. T1DM was associated with a significant increase in urinary excretion of both NGAL and MMP-9, and urine NGAL:Cr (NGAL corrected to urine creatinine) and urine MMP-9:Cr concentrations were highly correlated with each other. Both were also positively correlated with measurements of glycemic control and with albuminuria. Plasma MMP-9, urine MMP-9, and urine NGAL concentrations were significantly higher in females compared to males, and urine MMP-9:Cr concentrations displayed a menstrual cycle specific pattern. Increased urinary excretion of NGAL and MMP-9 supports a role for NGAL/MMP-9 dysregulation in renal dysfunction; moreover, gender-specific differences could support a gender contribution to pathological mechanisms or susceptibility for the development of renal complications in diabetes mellitus.


Subject(s)
Acute-Phase Proteins/urine , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/metabolism , Lipocalins/urine , Matrix Metalloproteinase 9/urine , Proto-Oncogene Proteins/urine , Sex Characteristics , Acute-Phase Proteins/genetics , Adolescent , Adult , Age Factors , Albuminuria/metabolism , Albuminuria/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Diabetic Nephropathies/physiopathology , Female , Gene Expression/physiology , Humans , Kidney Function Tests , Lipocalin-2 , Lipocalins/genetics , Male , Matrix Metalloproteinase 9/blood , Matrix Metalloproteinase 9/genetics , Podocytes/physiology , Proto-Oncogene Proteins/genetics , Tissue Inhibitor of Metalloproteinase-1/blood , Young Adult
17.
Bone ; 46(2): 410-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19772956

ABSTRACT

Distraction osteogenesis (DO) is a process which induces direct new bone formation as a result of mechanical distraction. Tumor necrosis factor-alpha (TNF) is a cytokine that can modulate osteoblastogenesis. The direct effects of TNF on direct bone formation in rodents are hypothetically mediated through TNF receptor 1 and/or 2 (TNFR1/2) signaling. We utilized a unique model of mouse DO to assess the effects of 1) TNFR homozygous null gene alterations on direct bone formation and 2) rmTNF on wild type (WT), TNFR1(-/-) (R1KO), and TNR2(-/-) (R2KO) mice. Radiological and histological analyses of direct bone formation in the distraction gaps demonstrated no significant differences between the WT, R1KO, R2KO, or TNFR1(-/-) and R2(-/-) (R1 and 2KO) mice. R1 and 2KO mice had elevated levels of serum TNF but demonstrated no inhibition of new bone formation. Systemic administration by osmotic pump of rmTNF during DO (10 microg/kg/day) resulted in significant inhibition of gap bone formation measures in WT and R2KO mice, but not in R1KO mice. We conclude that exogenous rmTNF and/or endogenous TNF act to inhibit new bone formation during DO by signaling primarily through TNFR1.


Subject(s)
Osteogenesis, Distraction , Osteogenesis/physiology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Tumor Necrosis Factor-alpha/pharmacology , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/pathology , Male , Mice , Osteogenesis/drug effects , Radiography , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Proteins/pharmacology , Staining and Labeling , Tumor Necrosis Factor-alpha/blood
18.
J Bone Miner Res ; 25(1): 114-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19580462

ABSTRACT

Skeletal changes accompanying aging are associated with both increased risk of fractures and impaired fracture healing, which, in turn, is due to compromised bone regeneration potential. These changes are associated with increased serum levels of selected proinflammatory cytokines, e.g., tumor necrosis factor alpha (TNF-alpha). We have used a unique model of bone regeneration to demonstrate (1) that aged-related deficits in direct bone formation can be restored to young mice by treatment with TNF blockers and (2) that the cyclin-dependent kinase inhibitor p21 is a candidate for mediation of the osteoinhibitory effects of TNF. It has been hypothesized recently that TNF antagonists may represent novel anabolic agents, and we believe that the data presented here represent a successful test of this hypothesis.


Subject(s)
Aging/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Regeneration/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Aging/blood , Animals , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cytokines/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiography , Receptors, Tumor Necrosis Factor, Type I/pharmacology , Receptors, Tumor Necrosis Factor, Type II/pharmacology , Recombinant Proteins/pharmacology , Solubility/drug effects , Tibia/diagnostic imaging , Tibia/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Wound Healing/drug effects
19.
Endocrinology ; 149(4): 1697-704, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18162513

ABSTRACT

Type 1 diabetes mellitus is associated with a number of disorders of skeletal health, conditions that rely, in part, on dynamic bone formation. A mouse model of distraction osteogenesis was used to study the consequences of streptozotocin-induced diabetes and insulin treatment on bone formation and osteoblastogenesis. In diabetic mice compared with control mice, new bone formation was decreased, and adipogenesis was increased in and around, respectively, the distraction gaps. Although insulin treatment restored bone formation to levels observed in nondiabetic control mice, it failed to significantly decrease adipogenesis. Molecular events altered during de novo bone formation in untreated type 1 diabetes mellitus, yet restored with insulin treatment were examined so as to clarify specific osteogenic genes that may contribute to diabetic bone disease. RNA from distraction gaps was analyzed by gene microarray and quantitative RT-PCR for osteogenic genes of interest. Runt-related transcription factor 2 (RUNX2), and several RUNX2 target genes, including matrix metalloproteinase-9, Akp2, integrin binding sialoprotein, Dmp1, Col1a2, Phex, Vdr, osteocalcin, and osterix, were all significantly down-regulated in the insulin-deficient, hyperglycemic diabetic animals; however, insulin treatment of diabetic animals significantly restored their expression. Expression of bone morphogenic protein-2, transcriptional coactivator with PDZ-binding motif, and TWIST2, all important regulators of RUNX2, were not impacted by the diabetic condition, suggesting that the defect in osteogenesis resides at the level of RUNX2 expression and its activity. Together, these data demonstrate that insulin and/or glycemic status can regulate osteogenesis in vivo, and systemic insulin therapy can, in large part, rescue the diabetic bone phenotype at the tissue and molecular level.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Diabetes Mellitus, Type 1/physiopathology , Osteogenesis/physiology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/genetics , Core Binding Factor Alpha 1 Subunit/physiology , Down-Regulation , Female , Insulin/pharmacology , Matrix Metalloproteinase 9/genetics , Mice , Osteogenesis, Distraction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics
20.
J Bone Miner Res ; 23(3): 338-49, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17967133

ABSTRACT

UNLABELLED: EtOH consumption significantly impaired anabolic rebuilding of bone after lactation. Lower BMD and BMC in EtOH-fed rats were associated with decreased bone formation in the proximal tibia, increased proportion of adipocytes, and increased expression of TNF-alpha. EtOH-induced skeletal deficits were prevented by treatment with either NAC or sTNFR1. These data suggest that postlactational anabolic rebuilding is influenced by EtOH consumption and may affect the long-term risk of osteopenia. INTRODUCTION: Despite significant loss of bone during lactation, BMD is restored by a powerful anabolic rebuilding process after weaning. A significant number of women resume alcohol consumption after weaning their offspring from breast feeding. The objectives of this study were to examine the consequences of chronic ethanol (EtOH) consumption on the postlactational rebuilding process and to investigate the underlying mechanisms by which EtOH mediates its detrimental effects. MATERIALS AND METHODS: Female Sprague-Dawley rats (n = 7-9 per group) were fed EtOH-containing diets (13 g/kg/d) for 1, 2, or 4 wk after weaning of their offspring. Skeletal parameters in the proximal tibia were examined using pQCT, microCT, and histomorphometric techniques, and interventional studies were performed on the mechanistic roles of EtOH-induced oxidative stress and TNF-alpha. RESULTS AND CONCLUSIONS: EtOH consumption completely abolished the anabolic bone rebuilding that occurred after lactation. Decreased BMD and BMC were associated with decreased bone formation and not with increased osteoclast activity. Furthermore, EtOH-fed rats showed greater proportion of fat volume/bone volume and expression of adipocyte-specific genes. EtOH-induced skeletal effects were mitigated by the dietary antioxidant, N-acetyl cysteine or by blocking TNF-alpha signaling. These data suggest EtOH consumption in the period immediately postweaning may significantly impair the mother's skeletal health and lead to long-term osteopenia.


Subject(s)
Alcoholism/metabolism , Bone Resorption/metabolism , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Lactation/metabolism , Osteogenesis/drug effects , Acetylcysteine/pharmacology , Adipocytes/metabolism , Adipocytes/pathology , Alcoholism/pathology , Animals , Bone Density/drug effects , Bone Diseases, Metabolic/chemically induced , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone Resorption/chemically induced , Bone Resorption/pathology , Female , Free Radical Scavengers/pharmacology , Oxidative Stress/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Risk Factors , Signal Transduction/drug effects , Tibia/metabolism , Tibia/pathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...