Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Nutr Biochem ; 34: 73-82, 2016 08.
Article in English | MEDLINE | ID: mdl-27239754

ABSTRACT

Nutrition is an important determinant of bone health and attainment of peak bone mass. Diets containing dried plum (DP) have been shown to increase bone volume and strength. These effects may be linked to the immune system and DP-specific polyphenols. To better understand these relationships, we studied DP in skeletally mature (6-month-old) and growing (1- and 2-month-old) C57Bl/6 male mice. In adult mice, DP rapidly (<2 weeks) increased bone volume (+32%) and trabecular thickness (+24%). These changes were associated with decreased osteoclast surface (Oc.S/BS) and decreased serum CTX, a marker of bone resorption. The reduction in Oc.S/BS was associated with a reduction in the osteoclast precursor pool. Osteoblast surface (Ob.S/BS) and bone formation rate were also decreased suggesting that the gain in bone in adult mice is a consequence of diminished bone resorption and formation, but resorption is reduced more than formation. The effects of DP on bone were accompanied by a decline in interleukins, TNF and MCP-1, suggesting that DP is acting in part through the immune system to suppress inflammatory activity and reduce the size of the osteoclast precursor pool. Feeding DP was accompanied by an increase in plasma phenolics, some of which have been shown to stimulate bone accrual. In growing and young adult mice DP at levels as low as 5% of diet (w/w) increased bone volume. At higher levels (DP 25%), bone volume was increased by as much as 94%. These data demonstrate that DP feeding dramatically increases peak bone mass during growth.


Subject(s)
Bone Development , Bone Resorption/prevention & control , Cytokines/antagonists & inhibitors , Food, Preserved , Fruit , Functional Food , Prunus domestica , Animals , Biomarkers/blood , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Remodeling , Bone Resorption/immunology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/cytology , Bone and Bones/immunology , Bone and Bones/metabolism , Bone and Bones/pathology , Cells, Cultured , Collagen Type I/blood , Cytokines/blood , Gene Expression Regulation, Developmental , Male , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/immunology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/cytology , Osteoclasts/immunology , Osteoclasts/metabolism , Osteoclasts/pathology , Peptide Fragments/blood , Peptides/blood
2.
Ther Adv Musculoskelet Dis ; 8(1): 15-27, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26834847

ABSTRACT

The lifespan of men is increasing and this is associated with an increased prevalence of osteoporosis in men. Osteoporosis increases the risk of bone fracture. Fractures are associated with increased disability and mortality, and public health problems. We review here the study of osteoporosis in men as obtained from a longitudinal cohort of community-based older men, the Osteoporotic Fractures in Men Study (MrOS).

3.
J Interferon Cytokine Res ; 35(6): 480-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25734366

ABSTRACT

Exposure to ionizing radiation can cause rapid mineral loss and increase bone-resorbing osteoclasts within metabolically active, cancellous bone tissue leading to structural deficits. To better understand mechanisms involved in rapid, radiation-induced bone loss, we determined the influence of total body irradiation on expression of select cytokines known both to stimulate osteoclastogenesis and contribute to inflammatory bone disease. Adult (16 week), male C57BL/6J mice were exposed to either 2 Gy gamma rays ((137)Cs, 0.8 Gy/min) or heavy ions ((56)Fe, 600MeV, 0.50-1.1 Gy/min); this dose corresponds to either a single fraction of radiotherapy (typical total dose is ≥10 Gy) or accumulates over long-duration interplanetary missions. Serum, marrow, and mineralized tissue were harvested 4 h-7 days later. Gamma irradiation caused a prompt (2.6-fold within 4 h) and persistent (peaking at 4.1-fold within 1 day) rise in the expression of the obligate osteoclastogenic cytokine, receptor activator of nuclear factor kappa-B ligand (Rankl), within marrow cells over controls. Similarly, Rankl expression peaked in marrow cells within 3 days of iron exposure (9.2-fold). Changes in Rankl expression induced by gamma irradiation preceded and overlapped with a rise in expression of other pro-osteoclastic cytokines in marrow (eg, monocyte chemotactic protein-1 increased by 11.9-fold, and tumor necrosis factor-alpha increased by 1.7-fold over controls). The ratio, Rankl/Opg, in marrow increased by 1.8-fold, a net pro-resorption balance. In the marrow, expression of the antioxidant transcription factor, Nfe2l2, strongly correlated with expression levels of Nfatc1, Csf1, Tnf, and Rankl. Radiation exposure increased a serum marker of bone resorption (tartrate-resistant acid phosphatase) and led to cancellous bone loss (16% decrement after 1 week). We conclude that total body irradiation (gamma or heavy-ion) caused temporal elevations in the concentrations of specific genes expressed within marrow and mineralized tissue related to bone resorption, including select cytokines that lead to osteoclastogenesis and elevated resorption; this is likely to account for rapid and progressive deterioration of cancellous microarchitecture following exposure to ionizing radiation.


Subject(s)
Bone Marrow/radiation effects , Bone Resorption/genetics , Bone and Bones/radiation effects , Gamma Rays/adverse effects , Osteoclasts/radiation effects , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Regulation , Isoenzymes/genetics , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Signal Transduction , Tartrate-Resistant Acid Phosphatase , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Whole-Body Irradiation
4.
J Orthop Res ; 32(2): 183-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24136593

ABSTRACT

Severe soft tissue trauma is associated with heterotopic ossification (HO), the abnormal deposition of bone at extra-skeletal sites. The pathophysiology of the development of trauma-induced HO remains largely unknown due in part to the lack of appropriate animal models. In this study, we sought to develop a new trauma-induced HO mouse model using muscle impact injury combined with low dose BMP-2. BMP-2 at doses ranging from 0 to 2 µg was injected into quadriceps muscles of adult male C57/BL6 mice. Animals then received a one-time quadriceps impaction injury to mimic the trauma associated with severe injuries. HO was monitored using in vivo microCT scanning at 1, 2, 4, and 8 weeks after treatment. After trauma, the expression of BMP-2, -4, BMP receptor 1, SOX9 and RUNX2 were increased in muscle. Although little or no HO was observed in mice receiving 1 µg BMP-2, combining this dose with muscle trauma produced an abundance of HO. At higher doses of BMP-2, trauma did not augment mineral deposition. These results suggest that BMP-2 signaling can sensitize muscle to trauma-induced HO. They also provide the basis for a new model to study the pathogenesis of trauma-induced HO.


Subject(s)
Disease Models, Animal , Ossification, Heterotopic/etiology , Quadriceps Muscle/injuries , Animals , Bone Morphogenetic Protein 2 , Bone and Bones/metabolism , Male , Mice , Mice, Inbred C57BL , Ossification, Heterotopic/chemically induced , Quadriceps Muscle/drug effects , Quadriceps Muscle/metabolism
5.
J Acad Nutr Diet ; 113(7): 928-35, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23706353

ABSTRACT

Diet plays a critical role in the pathogenesis of major chronic diseases common in populations of US veterans. The role of nutrition-focused wellness coaching in improving dietary behavior and/or reducing weight in overweight and obese US veterans is not known. At the San Francisco Veterans Affairs Medical Center, US veterans aged 25 to 80 years were randomized to receive nutrition coaching on eating behaviors at baseline only (control group, n=22) or an additional eight times over the course of 6 months (intervention group, n=28) in 2010-2011. Multiple coaching contacts decreased intake of energy, fat, and carbohydrate by 31% (P≤0.001) as evaluated by the 2005 Block food frequency questionnaire, which is composed of 111 food items. A weight loss of 5% from baseline (92.8 to 88.2 kg; P<0.01) was observed in the intervention group with mean body mass index decreasing from 30.4 to 28.9 (P<0.05). The control group showed a decrease in fat intake by 20% (P=0.01), but no statistically significant changes in intake of other nutrients or body weight (88.7 to 87.4 kg). Those in the intervention group reported diets at follow-up that were lower in cholesterol, saturated fat, sodium, sugar (P≤0.01), calcium (P< 0.05), and vitamin D (P<0.01), although when adjusted for energy (ie, nutrient density) calcium intake increased and vitamin D remained unchanged. Veterans' readiness to change eating behavior for weight loss improved with nutrition coaching. This study demonstrates that intermittent nutrition coaching can be an effective strategy to promote reductions in energy intake, body weight, and body mass index in overweight US veterans. Further research is needed to determine whether nutrition coaching improves other clinical outcomes and sustains weight loss.


Subject(s)
Diet, Fat-Restricted , Feeding Behavior , Health Promotion/methods , Nutritional Status , Veterans , Weight Loss , Body Mass Index , Calcium, Dietary/administration & dosage , Diet , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Energy Intake , Female , Follow-Up Studies , Food, Organic , Humans , Male , Middle Aged , San Francisco , Sodium, Dietary/administration & dosage , Surveys and Questionnaires , Vitamin D/administration & dosage
6.
FASEB J ; 27(9): 3505-13, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23704087

ABSTRACT

The chemokine CXCL12 and its receptor CXCR4 play a key role in regulation of hematopoietic stem cells and cell migratory function during morphogenesis. Osteoblasts express both the ligand and the receptor, but little is known about the role of CXCL12-CXCR4 signaling in maintaining skeletal homeostasis. Using Cre-Lox technology to delete CXCR4 in mature osteoblasts in mice, we show here a significant decrease in bone mass and alterations in cancellous bone structure. CXCR4 gene ablation increased the number of colony-forming units (CFU), CFU-positive for alkaline phosphatase (CFU-AP(+)), and mineralizing nodules in bone marrow stromal cell (BMSC) cultures. The adipocyte precursor population decreased in BMSCs harvested from the KO animals. The nonadherent population of BMSCs harvested from the long bone diaphysis of KO animals formed more osteoclasts, a finding that was associated with increased circulatory levels of pyridinoline, a marker of bone resorption. Our data show that osteoblast-specific CXCR4 deletion has profound effects on the mesenchymal stem cell pool and allocation to the osteoblastic and adipocytic cell lineages. They also show that CXCL12/CXCR4 signaling in the mature osteoblast can feedback to regulate the osteoclast precursor pool size and play a multifunctional role in regulating bone formation and resorption.


Subject(s)
Chemokine CXCL12/metabolism , Mesenchymal Stem Cells/cytology , Osteoclasts/cytology , Receptors, CXCR4/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL12/pharmacology , Genotype , Immunohistochemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Osteoclasts/drug effects , Osteoclasts/metabolism , Receptors, CXCR4/genetics , X-Ray Microtomography
7.
Bone ; 52(1): 424-32, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23088940

ABSTRACT

INTRODUCTION: While the anti-resorptive effects of the bisphosphonates (BPs) are well documented, many questions remain about their mechanisms of action, particularly following long-term use. This study evaluated the effects of alendronate (Ale) treatment on TGF-ß1 signaling in mesenchymal stem cells (MSCs) and osteocytes, and the relationship between prolonged alendronate treatment on systemic TGF-ß1 levels and bone strength. METHODS: TGF-ß1 expression and signaling were evaluated in MSCs and osteocytic MLO-Y4 cells following Ale treatment. Serum total TGF-ß1 levels, a bone resorption marker (DPD/Cr), three-dimensional microCT scans and biomechanical tests from both the trabecular and cortical bone were measured in ovariectomized rats that either received continuous Ale treatment for 360 days or Ale treatment for 120 days followed by 240 days of vehicle. Linear regression tests were performed to determine the association of serum total TGF-ß1 levels and both the trabecular (vertebrae) and cortical (tibiae) bone strength. RESULTS: Ale increased TGF-ß1 signaling in the MSCs but not in the MLO-Y4 cells. Ale treatment increased serum TGF-ß1 levels and the numbers of TGF-ß1-positive osteocytes and periosteal cells in cortical bone. Serum TGF-ß1 levels were not associated with vertebral maximum load and strength but was negatively associated with cortical bone maximum load and ultimate strength. CONCLUSIONS: The increase of serum TGF-ß1 levels during acute phase of estrogen deficiency is likely due to increased osteoclast-mediated release of matrix-derived latent TGF-ß1. Long-term estrogen-deficiency generally results in a decline in serum TGF-ß1 levels that are maintained by Ale treatment. Measuring serum total TGF-ß1 levels may help to determine cortical bone quality following alendronate treatment.


Subject(s)
Alendronate/therapeutic use , Bone Density Conservation Agents/therapeutic use , Bone and Bones/physiology , Estrogens/deficiency , Models, Animal , Transforming Growth Factor beta1/blood , Animals , Biomechanical Phenomena , Female , Immunohistochemistry , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Signal Transduction
8.
Am J Physiol Endocrinol Metab ; 303(11): E1354-62, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23047986

ABSTRACT

Loss of skeletal weight bearing or skeletal unloading as occurs during spaceflight inhibits bone formation and stimulates bone resorption. These are associated with a decline in the osteoblast (Ob.S/BS) and an increase in the osteoclast (Oc.S/BS) bone surfaces. To determine the temporal relationship between changes in the bone cells and their marrow precursor pools during sustained unloading, and whether genetic background influences these relationships, we used the hindlimb unloading model to induce bone loss in two strains of mice known to respond to load and having significantly different cancellous bone volumes (C57BL/6 and DBA/2 male mice). Skeletal unloading caused a progressive decline in bone volume that was accompanied by strain-specific changes in Ob.S/BS and Oc.S/BS. These were associated with a sustained reduction in the osteoprogenitor population and a dramatic but transient increase in the osteoclast precursor pool size in both strains. The results reveal that bone adaptation to skeletal unloading involves similar rapid changes in the osteoblast and osteoclast progenitor populations in both strains of mice but striking differences in Oc.S/BS dynamics, BFR, and cancellous bone structure. These strain-specific differences suggest that genetics plays an important role in determining the osteoblast and osteoclast populations on the bone surface and the dynamics of bone loss in response to skeletal unloading.


Subject(s)
Bone Remodeling/physiology , Bone Resorption/pathology , Bone and Bones/cytology , Hindlimb Suspension/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Animals , Bone Marrow Cells/cytology , Bone Resorption/physiopathology , Bone and Bones/pathology , Cell Differentiation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Space Flight , Space Simulation , Species Specificity , Stem Cells/cytology
9.
Proc Natl Acad Sci U S A ; 109(35): 14092-7, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22886088

ABSTRACT

The Wnt antagonist Sost has emerged as a key regulator of bone homeostasis through the modulation of Lrp4/5/6 Wnt coreceptors. In humans, lack of Sclerostin causes sclerosteosis and van Buchem (VB) disease, two generalized skeletal hyperostosis disorders that result from hyperactive Wnt signaling. Unlike sclerosteosis, VB patients lack SOST coding mutations but carry a homozygous 52 kb noncoding deletion that is essential for the transcriptional activation of SOST in bone. We recently identified a putative bone enhancer, ECR5, in the VB deletion region, and showed that the transcriptional activity of ECR5 is controlled by Mef2C transcription factor in vitro. Here we report that mice lacking ECR5 or Mef2C through Col1-Cre osteoblast/osteocyte-specific ablation result in high bone mass (HBM) due to elevated bone formation rates. We conclude that the absence of the Sost-specific long-range regulatory element ECR5 causes VB disease in rodents, and that Mef2C is the main transcription factor responsible for ECR5-dependent Sost transcriptional activation in the adult skeleton.


Subject(s)
Bone Remodeling/genetics , Enhancer Elements, Genetic/genetics , Glycoproteins/genetics , Hyperostosis/genetics , Myogenic Regulatory Factors/genetics , Osteocytes/physiology , Syndactyly/genetics , Adaptor Proteins, Signal Transducing , Age Factors , Animals , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Female , Femur/cytology , Femur/physiology , Gene Deletion , Glycoproteins/metabolism , Hyperostosis/metabolism , Intercellular Signaling Peptides and Proteins , Lac Operon , MEF2 Transcription Factors , Male , Mandible/abnormalities , Mandible/metabolism , Mice , Mice, Transgenic , Myogenic Regulatory Factors/metabolism , Osteochondrodysplasias , Osteosclerosis/genetics , Osteosclerosis/metabolism , Signal Transduction/genetics , Skull/abnormalities , Skull/metabolism , Syndactyly/metabolism , Transcriptional Activation/genetics
10.
Calcif Tissue Int ; 91(1): 50-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22644321

ABSTRACT

Sclerostin functions as an antagonist to Wnt signaling and inhibits bone-forming activity. We studied the effects of skeletal unloading and treatment with sclerostin antibody (Scl-Ab) on mesenchymal stem cell, osteoprogenitor and osteoclast precursor pools, and their relationship to bone formation and resorption. Male C57BL/6 mice (5-months-old) were hind limb unloaded for 1 week or allowed normal ambulation and treated with Scl-Ab (25 mg/kg, s.c. injections on days 1 and 4) or placebo. Unloading decreased the serum concentration of bone formation marker P1NP (-35 %), number of colony-forming units (CFU) (-38 %), alkaline phosphatase-positive CFUs (CFU-AP+) (-51 %), and calcified nodules (-35 %); and resulted in a fourfold increase in the number of osteoclast precursors. The effects of Scl-Ab treatment on unloaded and normally loaded mice were nearly identical; Scl-Ab increased serum P1NP and the number of CFU, CFU-AP+, and calcified nodules in ex vivo cultures; and increased osteoblast and bone mineralizing surfaces in vivo. Although the marrow-derived osteoclast precursor population increased with Scl-Ab, the bone osteoclast surface did not change, and the serum concentration of osteoclast activity marker TRACP5b decreased. Our data suggest that short-term Scl-Ab treatment can prevent the decrease in osteoprogenitor population associated with skeletal unloading and increase osteoblast surface and bone mineralizing surface in unloaded animals. The anabolic effects of Scl-Ab treatment on bone are preserved during skeletal unloading. These findings suggest that Scl-Ab treatment can both increase bone formation and decrease bone resorption, and provide a new means for prevention and treatment of disuse osteoporosis.


Subject(s)
Antibodies/metabolism , Bone Marrow/metabolism , Glycoproteins/immunology , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Resorption , Glycoproteins/genetics , Glycoproteins/metabolism , Hindlimb Suspension , Intercellular Signaling Peptides and Proteins , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tartrate-Resistant Acid Phosphatase
11.
Nat Med ; 18(3): 456-62, 2012 Feb 05.
Article in English | MEDLINE | ID: mdl-22306732

ABSTRACT

Aging reduces the number of mesenchymal stem cells (MSCs) that can differentiate into osteoblasts in the bone marrow, which leads to impairment of osteogenesis. However, if MSCs could be directed toward osteogenic differentiation, they could be a viable therapeutic option for bone regeneration. We have developed a method to direct MSCs to the bone surface by attaching a synthetic high-affinity and specific peptidomimetic ligand (LLP2A) against integrin α4ß1 on the MSC surface to a bisphosphonate (alendronate, Ale) that has a high affinity for bone. LLP2A-Ale induced MSC migration and osteogenic differentiation in vitro. A single intravenous injection of LLP2A-Ale increased trabecular bone formation and bone mass in both xenotransplantation studies and in immunocompetent mice. Additionally, LLP2A-Ale prevented trabecular bone loss after peak bone acquisition was achieved or as a result of estrogen deficiency. These results provide proof of principle that LLP2A-Ale can direct MSCs to the bone to form new bone and increase bone strength.


Subject(s)
Bone Regeneration/drug effects , Dipeptides/pharmacology , Mesenchymal Stem Cells/cytology , Osteoblasts/physiology , Osteogenesis/drug effects , Peptidomimetics/pharmacokinetics , Phenylurea Compounds/pharmacology , Alendronate/analogs & derivatives , Alendronate/chemical synthesis , Alendronate/pharmacology , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cell Migration Assays , Cell Proliferation/drug effects , Cells, Cultured , Dipeptides/chemical synthesis , Humans , Immunocompetence/genetics , Integrin alpha4beta1/chemistry , Integrin alpha4beta1/metabolism , Mice , Osteoblasts/cytology , Peptidomimetics/chemical synthesis , Phenylurea Compounds/chemical synthesis , Transplantation, Heterologous
12.
Bone ; 50(3): 628-37, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22154841

ABSTRACT

We examined age-related changes in biochemical markers and regulators of osteoblast and osteoclast activity in C57BL/6 mice to assess their utility in explaining age-related changes in bone. Several recently discovered regulators of osteoclasts and osteoblasts were also measured to assess concordance between their systemic levels versus their levels in marrow plasma, to which bone cells are directly exposed. MicroCT of 6-, 12-, and 24-month-old mice indicated an early age-related loss of trabecular bone volume and surface, followed by endocortical bone loss and periosteal expansion. Trabecular bone loss temporally correlated with reductions in biomarkers of bone formation and resorption in both peripheral blood and bone marrow. Endocortical bone loss and periosteal bone gain were not reflected in these protein biomarkers, but were well correlated with increased expression of osteocalcin, rank, tracp5b, and cathepsinK in RNA extracted from cortical bone. While age-related changes in bone turnover markers remained concordant in blood versus marrow, aging led to divergent changes in blood versus marrow for the bone cell regulators RANKL, OPG, sclerostin, DKK1, and serotonin. Bone expression of runx2 and osterix increased progressively with aging and was associated with an increase in the number of osteoprogenitors and osteoclast precursors. In summary, levels of biochemical markers of bone turnover in blood and bone marrow plasma were predictive of an age-related loss of trabecular surfaces in adult C57BL/6 mice, but did not predict gains in cortical surfaces resulting from cortical expansion. Unlike these turnover markers, a panel of bone cell regulatory proteins exhibited divergent age-related changes in marrow versus peripheral blood, suggesting that their circulating levels may not reflect local levels to which osteoclasts and osteoblasts are directly exposed.


Subject(s)
Aging/metabolism , Bone Remodeling/physiology , Bone and Bones/metabolism , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Animals , Biomarkers/blood , Bone Marrow/metabolism , Cells, Cultured , Male , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism
13.
FASEB J ; 25(10): 3366-76, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21705669

ABSTRACT

In response to cellular insult, several pathways can be activated, including necrosis, apoptosis, and autophagy. Because glucocorticoids (GCs) have been shown to induce both osteocyte apoptosis and autophagy, we sought to determine whether osteocyte cell fate in the presence of GCs was dose dependent by performing in vivo and in vitro studies. Male Swiss-Webster mice were treated with slow-release prednisolone pellets at 1.4, 2.8, and 5.6 mg/kg/d for 28 d. An osteocyte cell line, MLO-Y4 cells, was treated with various doses of dexamethasone. We found that GC treatments dose dependently decreased activation of antioxidant-, autophagy-, and antiapoptosis-focused RT-PCR gene pathways in mouse cortical bone. The activation of antioxidant genes was correlated with autophagy gene expression after the GC treatments. The presence of osteocyte autophagy, as detected by immunostaining for LC3, increased ∼50% at the distal femur cortical bone region but not at trabecular bone region at the 1.4 and 2.8 mg/kg/d GC dose levels. The number of apoptotic osteocytes was increased at the cortical bone region by ∼40% initially observed at the 2.8 mg/kg/d dose level. In addition, the presence of the osteocyte autophagy was associated with an increased protein level of cathepsin K in vitro after the GC treatments. In summary, we found that GC treatment dose-dependently decreased antioxidant gene expression, with lower GC doses activating autophagy, whereas a higher dose increased apoptosis. These data suggest that autophagy may provide a mechanism for osteocytes to survive the stress after GC exposure and provide further insight into how GCs alter bone cell fate.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Osteocytes/drug effects , Prednisolone/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cathepsin K/metabolism , Cell Line , Delayed-Action Preparations , Dexamethasone/administration & dosage , Dose-Response Relationship, Drug , Glucocorticoids/administration & dosage , Male , Mice , Osteocytes/metabolism , Prednisolone/administration & dosage
14.
J Bone Miner Res ; 26(3): 569-81, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20839286

ABSTRACT

Osteoporotic patients treated with antiresorptive or anabolic agents experience an increase in bone mass and a reduction in incident fractures. However, the effects of these medications on bone quality and strength after a prolonged discontinuation of treatment are not known. We evaluated these effects in an osteoporotic rat model. Six-month-old ovariectomized (OVX) rats were treated with placebo, alendronate (ALN, 2 µg/kg), parathyroid hormone [PTH(1-34); 20 µg/kg], or raloxifene (RAL, 2 mg/kg) three times a week for 4 months and withdrawn from the treatments for 8 months. Treatment with ALN, PTH, and RAL increased the vertebral trabecular bone volume (BV/TV) by 47%, 53%, and 31%, with corresponding increases in vertebral compression load by 27%, 51%, and 31%, respectively (p < .001). The resulting bone strength was similar to that of the sham-OVX control group with ALN and RAL and higher (p < .001) with PTH treatment. After 4 months of withdrawal, bone turnover (BFR/BS) remained suppressed in the ALN group versus the OVX controls (p < .001). The vertebral strength was higher than in the OVX group only in ALN-treated group (p < .05), whereas only the PTH-treated animals showed a higher maximum load in tibial bending versus the OVX controls (p < .05). The vertebral BV/TV returned to the OVX group level in both the PTH and RAL groups 4 months after withdrawal but remained 25% higher than the OVX controls up to 8 months after withdrawal of ALN (p < .05). Interestingly, cortical bone mineral density increased only with PTH treatment (p < .05) but was not different among the experimental groups after withdrawal. At 8 months after treatment withdrawal, none of the treatment groups was different from the OVX control group for cortical or cancellous bone strength. In summary, both ALN and PTH maintained bone strength (maximum load) 4 months after discontinuation of treatment despite changes in bone mass and bone turnover; however, PTH maintained cortical bone strength, whereas ALN maintained cancellous bone strength. Additional studies on the long-term effects on bone strength after discontinuation and with combination of osteoporosis medications are needed to improve our treatment of osteoporosis.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone and Bones/drug effects , Bone and Bones/physiology , Amino Acids/urine , Animals , Biomechanical Phenomena/drug effects , Bone Density Conservation Agents/administration & dosage , Bone and Bones/diagnostic imaging , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/physiology , Ovariectomy , Rats , Rats, Sprague-Dawley , Spine/diagnostic imaging , Spine/drug effects , Spine/physiology , Tibia/diagnostic imaging , Tibia/drug effects , Tibia/physiology , Weight-Bearing/physiology , X-Ray Microtomography
15.
PLoS One ; 5(7): e11410, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20625385

ABSTRACT

Augmentation of the peak bone mass (PBM) may be one of the most effective interventions to reduce the risk of developing osteoporosis later in life; however treatments to augment PBM are currently limited. Our study evaluated whether a greater PBM could be achieved either in the progesterone nuclear receptor knockout mice (PRKO) or by using a nuclear progesterone receptor (nPR) antagonist, RU486 in mice. Compared to their wild type (WT) littermates the female PRKO mice developed significantly higher cancellous and cortical mass in the distal femurs, and this was associated with increased bone formation. The high bone mass phenotype was partially reproduced by administering RU486 in female WT mice from 1-3 months of age. Our results suggest that the inhibition of the nPR during the rapid bone growth period (1-3 months) increases osteogenesis, which results in acquisition of higher bone mass. Our findings suggest a crucial role for progesterone signaling in bone acquisition and inhibition of the nPR as a novel approach to augment bone mass, which may have the potential to reduce the burden of osteoporosis.


Subject(s)
Bone Density/physiology , Bone Development/drug effects , Osteoblasts/metabolism , Receptors, Progesterone/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Body Weight/drug effects , Body Weight/genetics , Bone Density/drug effects , Bone Density/genetics , Bone Development/genetics , Bone Resorption/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Female , Hormone Antagonists/pharmacology , Male , Mice , Mice, Knockout , Mifepristone/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Sex Factors
16.
Am J Physiol Renal Physiol ; 298(6): F1315-22, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20200094

ABSTRACT

Calcimimetics activate the calcium-sensing receptor (CaR) and reduce parathyroid hormone (PTH) by increasing the sensitivity of the parathyroid CaR to ambient calcium. The calcimimetic, cinacalcet, is effective in treating secondary hyperparathyroidism in dialysis patients [chronic kidney disease (CKD 5)], but little is known about its effects on stage 3-4 CKD patients. We compared cinacalcet and paricalcitol in uremic rats with creatinine clearances "equivalent" to patients with CKD 3-4. Uremia was induced in anesthetized rats using the 5/6th nephrectomy model. Groups were 1) uremic control, 2) uremic + cinacalcet (U+Cin; 15 mg x kg(-1) x day(-1) po for 6 wk), 3) uremic + paricalcitol (U+Par; 0.16 microg/kg, 3 x wk, ip for 6 wk), and 4) normal. Unlike U+Par animals, cinacalcet promoted hypocalcemia and marked hyperphosphatemia. The Ca x P in U+Cin rats was twice that of U+Par rats. Both compounds suppressed PTH. Serum 1,25-(OH)(2)D(3) was decreased in both U+Par and U+Cin rats. Serum FGF-23 was increased in U+Par but not in U+Cin, where it tended to decrease. Analysis of tibiae showed that U+Cin, but not U+Par, rats had reduced bone volume. U+Cin rats had similar bone formation and reduced osteoid surface, but higher bone resorption. Hypocalcemia, hyperphosphatemia, low 1,25-(OH)(2)D(3), and cinacalcet itself may play a role in the detrimental effects on bone seen in U+Cin rats. This requires further investigation. In conclusion, due to its effects on bone and to the hypocalcemia and severe hyperphosphatemia it induces, we believe that cinacalcet should not be used in patients with CKD without further detailed studies.


Subject(s)
Ergocalciferols/pharmacology , Fibroblast Growth Factors/blood , Hyperparathyroidism/drug therapy , Naphthalenes/pharmacology , Phosphorus/blood , Renal Insufficiency, Chronic/drug therapy , Tibia/drug effects , Animals , Biomarkers/blood , Biomarkers/urine , Bone Resorption/chemically induced , Bone Resorption/metabolism , Calcitriol/blood , Calcium/urine , Cinacalcet , Creatinine/blood , Disease Models, Animal , Ergocalciferols/adverse effects , Female , Hyperparathyroidism/metabolism , Hyperparathyroidism/pathology , Hyperphosphatemia/chemically induced , Hyperphosphatemia/metabolism , Hypocalcemia/chemically induced , Hypocalcemia/metabolism , Naphthalenes/adverse effects , Nephrectomy , Parathyroid Hormone/blood , Phosphorus/urine , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Severity of Illness Index , Tibia/metabolism , Tibia/pathology , Uremia/drug therapy , Uremia/metabolism
17.
Bone ; 46(5): 1238-43, 2010 May.
Article in English | MEDLINE | ID: mdl-20149909

ABSTRACT

Techniques for assessing bone dynamic are in high demand. Calcium (Ca) kinetic studies are currently being used in our clinical studies of bone turnover in adolescents and elderly. The technique has rarely been compared to the standard method of bone dynamic histomorphometry. We perturbed bone turnover through ovariectomy and sub-optimal dietary Ca in a female rat model to cross-calibrate Ca kinetics against dynamic histomorphometry. Kinetic studies involved oral and intravenous administration of (45)Ca and monitoring the tracer in blood, urine, feces, and bone over a 3-day period as part of a metabolic Ca balance study. Histomorphometric indices of mineral apposition rate, mineralizing surface, and bone formation rate were obtained from proximal metaphysis and mid-diaphysis region of tibial bone. Bone mineralization and resorption rates at the whole skeletal level as evaluated by kinetic studies were significantly correlated with the volume-based bone formation rate (BFR/BV) evaluated by dynamic histomorphometry in metaphyseal trabecular bone (r=0.72 and r=0.61, respectively, p<0.001) and surface-based bone formation rate (BFR/BS) in tibial cortex (r=0.63, p<0.001 and r=0.59, p<0.01, respectively). Significant correlations were also demonstrated between bone resorption and mineralization rates at the whole skeletal level (r=0.91, p<0.001) using (45)Ca kinetic data. Ca kinetic modeling showed an increase (p<0.001) in skeletal resorption and formation rates in response to ovariectomy (27.6 vs. 13.8 mg/d for bone resorption and 42.7 vs. 28 mg/d for bone formation in ovariectomized vs. their Sham-operated control animals, respectively). Ca kinetic data also showed that bone formation decreased by 30% and whole bone balance by 50%, when dietary Ca level was reduced from 0.4% to 0.2% (34.2 vs. 23.8 mg/d and 10.4 vs. 5.1 mg/d, respectively, p<0.001). Our data suggest that Ca kinetic studies can be used reliably to rapidly detect changes in bone turnover at the whole skeletal level in response to interventions.


Subject(s)
Bone and Bones/metabolism , Calcium/metabolism , Animals , Bone Resorption/metabolism , Bone Resorption/physiopathology , Calcium, Dietary/metabolism , Female , Kinetics , Osteogenesis/physiology , Ovariectomy , Rats , Rats, Sprague-Dawley
18.
Bone ; 46(5): 1267-74, 2010 May.
Article in English | MEDLINE | ID: mdl-19931661

ABSTRACT

We report the results of a series of experiments designed to determine the effects of ibandronate (Ibn) and risedronate (Ris) on a number of bone quality parameters in aged osteopenic rats to explain how bone material and bone mass may be affected by the dose of bisphosphonates (BP) and contribute to their anti-fracture efficacy. Eighteen-month old female rats underwent either ovariectomy or sham surgery. The ovariectomized (OVX) groups were left untreated for 2 months to develop osteopenia. Treatments started at 20 months of age as follows: sham and OVX control (treated with saline), OVX + risedronate 30 and 90 (30 or 90 microg/kg/dose), and OVX + ibandronate 30 and 90 (30 or 90 microg/kg/dose). The treatments were given monthly for 4 months by subcutaneous injection. At sacrifice at 24 months of age the 4th lumbar vertebra was used for microCT scans (bone mass, architecture, and degree of mineralization of bone, DMB) and histomorphometry, and the 6th lumbar vertebra, tibia, and femur were collected for biomechanical testing to determine bone structural and material strength, cortical fracture toughness, and tissue elastic modulus. The compression testing of the vertebral bodies (LVB6) was simulated using finite-element analysis (FEA) to also estimate the bone structural stiffness. Both Ibn and Ris dose-dependently increased bone mass and improved vertebral bone microarchitecture and mechanical properties compared to OVX control. Estimates of vertebral maximum stress from FEA were correlated with vertebral maximum load (r=0.5, p<0.001) and maximum stress (r=0.4, p<0.005) measured experimentally. Tibial bone bending modulus and cortical strength increased compared to OVX with both BP but no dose-dependent effect was observed. DMB and elastic modulus of trabecular bone were improved with Ibn 30 compared to OVX but were not affected in other BP-treated groups. DMB of tibial cortical bone showed no change with BP treatments. The fracture toughness examined in midshaft femurs did not change with BP even with the higher doses. In summary, the anti-fracture efficacy of BP is largely due to their preservation of bone mass and while the higher doses further improve the bone structural properties do not improve the localized bone material characteristics such as tissue strength, elastic modulus, and cortical toughness.


Subject(s)
Aging/physiology , Bone Density Conservation Agents/pharmacology , Bone and Bones/drug effects , Bone and Bones/physiology , Diphosphonates/pharmacology , Animals , Bone Density Conservation Agents/therapeutic use , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/physiopathology , Bone and Bones/metabolism , Bone and Bones/physiopathology , Compressive Strength/drug effects , Diphosphonates/therapeutic use , Female , Finite Element Analysis , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/metabolism , Lumbar Vertebrae/physiology , Lumbar Vertebrae/physiopathology , Ovariectomy , Rats , Rats, Inbred F344
19.
J Bone Miner Res ; 25(2): 190-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19594295

ABSTRACT

Secreted frizzled-related protein 1 (sFRP1) is an antagonist of Wnt signaling, an important pathway in maintaining bone homeostasis. In this study we evaluated the skeletal phenotype of mice overexpressing sFRP1 (sFRP1 Tg) and the interaction of parathyroid hormone (PTH) treatment and sFRP1 (over)expression. Bone mass and microarchitecture were measured by micro-computed tomography (microCT). Osteoblastic and osteoclastic cell maturation and function were assessed in primary bone marrow cell cultures. Bone turnover was assessed by biochemical markers and dynamic bone histomorphometry. Real-time PCR was used to monitor the expression of several genes that regulate osteoblast maturation and function in whole bone. We found that trabecular bone mass measurements in distal femurs and lumbar vertebral bodies were 22% and 51% lower in female and 9% and 33% lower in male sFRP1 Tg mice, respectively, compared with wild-type (WT) controls at 3 months of age. Genes associated with osteoblast maturation and function, serum bone formation markers, and surface based bone formation were significantly decreased in sFRP1 Tg mice of both sexes. Bone resorption was similar between sFRP1 Tg and WT females and was higher in sFRP1 Tg male mice. Treatment with hPTH(1-34) (40 microg/kg/d) for 2 weeks increased trabecular bone volume in WT mice (females: +30% to 50%; males: +35% to 150%) compared with sFRP1 Tg mice (females: +5%; males: +18% to 54%). Percentage increases in bone formation also were lower in PTH-treated sFRP1 Tg mice compared with PTH-treated WT mice. In conclusion, overexpression of sFRP1 inhibited bone formation as well as attenuated PTH anabolic action on bone. The gender differences in the bone phenotype of the sFRP1 Tg animal warrants further investigation.


Subject(s)
Cell Differentiation , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/physiology , Parathyroid Hormone/pharmacology , Animals , Blotting, Western , Bone Resorption , Cell Differentiation/drug effects , Cells, Cultured , Female , Gene Expression , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
20.
J Nutr ; 139(10): 1908-13, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19710157

ABSTRACT

Soy isoflavones and their metabolites, with estrogenic activity, have been considered candidates for reducing postmenopausal bone loss. In this study, we examined the effect of dietary equol, a bioactive metabolite of the soy isoflavone daidzein, on equol tissue distribution, bone parameters, and reproductive tissue activity using an adult ovariectomized (OVX) rat model. An 8-wk feeding study was conducted to compare 4 dietary treatments of equol (0, 50, 100, 200 mg/kg diet) in 6-mo-old OVX female Sprague-Dawley rats. A dose response increase in tissue equol concentrations was observed for serum, liver, kidney, and heart, and a plateau occurred at 100 mg equol/kg diet for intestine. In OVX rats receiving 200 mg equol/kg diet, femoral calcium concentration was greater than those receiving lower doses but was still less than SHAM (P < 0.05), and other bone measures were not improved. Tibia calcium concentrations were lower in OVX rats receiving 100 and 200 mg equol/kg diet compared with the OVX control rats. Trabecular bone mineral density of tibia was also lower in equol-fed OVX rats. At this dietary equol intake, uterine weight was higher (P < 0.05) than in other OVX groups but lower than the SHAM-operated intact rats. The 200 mg/kg diet dose of dietary equol significantly increased proliferative index in the uterine epithelium. Dietary equol had no stimulatory effect on mammary gland epithelium. We conclude that in OVX rats, a dietary equol dose that had modest effect on bone also exerts mild uterotropic effects.


Subject(s)
Bone Density/drug effects , Dietary Supplements , Isoflavones/pharmacology , Phytoestrogens/pharmacology , Uterus/drug effects , Animals , Bone and Bones/chemistry , Calcium/analysis , Equol , Female , Lethal Dose 50 , Organ Size , Ovariectomy , Rats , Rats, Sprague-Dawley , Uterus/anatomy & histology
SELECTION OF CITATIONS
SEARCH DETAIL
...